Vous êtes sur la page 1sur 14

Pharmacological Research 130 (2018) 259–272

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Review

Tocotrienols: The promising analogues of vitamin E for cancer


therapeutics
Bethsebie Lalduhsaki Sailo, Kishore Banik, Ganesan Padmavathi, Monisha Javadi,
Devivasha Bordoloi, Ajaikumar B. Kunnumakkara ∗,1,2
Cancer Biology Laboratory and DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering,
Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India

a r t i c l e i n f o a b s t r a c t

Article history: Despite the significant advancements in the diagnosis and treatment of cancer, it still remains one of
Received 14 November 2017 the most fatal diseases in the world due to the lack of sensitive diagnosis methods and effective drugs.
Received in revised form 6 February 2018 Therefore, discovering novel therapies that are safe, efficacious and affordable are required for the bet-
Accepted 12 February 2018
ter management of this disease. Tocotrienols, analogues of vitamin E have gained increased attention
Available online 27 February 2018
due to their safety and efficacy. Extensive research over the past several years has strongly indicated
that tocotrienols can efficiently prevent/inhibit the growth of different cancers such as cancers of blood,
Keywords:
brain, breast, cervical, colon, liver, lung, pancreas, prostate, skin, stomach etc. This is mainly accredited
Tocotrienols
Cancer
to their ability to modulate various molecular targets involved in cancer cell proliferation, survival, inva-
Chemoprevention sion, angiogenesis, and metastasis such as NF-␬B, STAT3, Akt/mTOR, etc. In addition, increasing lines
Chemosensitization of evidence has shown that tocotrienols can sensitize cancer cells to chemotherapeutic agents such as
Molecular targets celecoxib, doxorubicin, erlotinib, gefitinib, gemcitabine, paclitaxel, statin etc. Moreover, several clinical
trials have confirmed the safety and tolerability of tocotrienols in humans. This review summarizes the
potential of tocotrienols for the prevention and treatment of different cancers based on the available
in vitro, in vivo and clinical studies.
© 2018 Published by Elsevier Ltd.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260
2. Vitamin E . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260
3. Tocotrienols are superior to tocopherols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260
4. Tocotrienols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260
5. Molecular targets of tocotrienols in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
6. Tocotrienols as chemopreventive and chemotherapeutic agents for various cancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
6.1. Breast cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
6.2. Colorectal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264
6.3. Gastric cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
6.4. Glioblastoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
6.5. Leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
6.6. Liver cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
6.7. Lung cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
6.8. Mesothelioma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265

∗ Corresponding author at: Cancer Biology Laboratory and DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengi-
neering, Indian Institute of Technology Guwahati, Room # 005, O-Block, Guwahati, Assam, 781039, India.
E-mail address: kunnumakkara@iitg.ernet.in (A.B. Kunnumakkara).
1
Websites: www.iitg.ernet.in/kunnumakkara.
2
www.iitg.ac.in/kunnumakkara.

https://doi.org/10.1016/j.phrs.2018.02.017
1043-6618/© 2018 Published by Elsevier Ltd.
260 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

6.9. Pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266


6.10. Prostate cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266
6.11. Skin cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266
6.12. Other cancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266
7. Chemosensitizing effect of tocotrienols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266
8. Clinical studies on tocotrienols- completed and ongoing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
9. Conclusions and future prospect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
Conflict of interests . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 269
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 269

1. Introduction higher anti-oxidant activity of T3 is attributed to their structures


that assist in their integration into the cell i.e. short tail, unsatu-
Despite extensive research over the last several decades for find- rated, with three double bonds, whereas T isoforms have saturated
ing a cure for cancer, it continues to remain one of the most fatal side-chain without double bonds (Fig. 1) [4,9]. Moreover, unlike
chronic diseases today. The incidence and mortality rates of can- T isoforms, T3 isoforms are predominant for their innate anti-
cer continue to rise, accounting for approximately 14.1 million cancer and chemosensitizing activities and confer their anti-cancer
new cases and 8.2 million deaths worldwide in 2012 alone [1]. activities independently from its antioxidant function [4,14]. In
Conventional treatment modalities for cancer including surgery, agreement with this, studies have delineated that T3 are more
radiation, and chemotherapy have failed to completely eradicate efficient than T in abrogating various cancer-promoting pathways
cancer. Hence, finding a cure for cancer is the holy grail of medical including 5-lipoxygenase (5-LOX)-catalyzed eicosanoids, cyclooxy-
research. In addition, most of the recently developed therapies are genase (COX), nuclear transcription factor-kappaB (NF-␬B) and
extremely expensive and not affordable to majority of the popula- signal transducer and activator of transcription factor 3 (STAT3),
tion. On account of the alarming increase in cancer incidence and which may be due to the higher absorption of T3 by cancer cells,
the spiraling cost of unsuccessful treatments, cancer prevention attributed to their unsaturated isoprenoid side chain that enhances
and treatment using nutraceuticals has recently become a subject their lipophilic property [15]. Moreover, it has been exhibited that
of immense interest amongst researchers [2]. Further, numerous T3 have greater potency than T in instigating apoptosis and are
studies have evinced that phytochemicals found in fruits, vegeta- found to preferentially accumulate in breast cancer cells as com-
bles, and spices evidently exhibit chemopreventive and therapeutic pared to T [16]. More importantly, it has been demonstrated that
properties against various tumors by modulating different types ␥- and ␦-T3 induced apoptosis in various tumors via distinct mech-
of oncogenic targets such as cytokines, chemokines, growth fac- anisms that are not shared by the T isoforms, such as induction of
tors, inflammatory enzymes, transcription factors etc. [3,4]. Thus, endoplasmic reticulum (ER) stress and increased translocation of
these compounds serve as profound sources of leads for cancer estrogen receptor beta (ER␤) into the nucleus by serving as a lig-
drug development. In line with this, a group of dietary compo- and for ER␤ [17]. Notably, growing body evidence has revealed T3
nents, known as tocotrienols (T3)-analogues of vitamin E have isoforms as potent chemosensitizers, but not T isoforms, thus fur-
entered the limelight in the field of cancer drug development [5–7]. ther validating its higher potency than T. Considering these aspects,
Tocotrienols (T3) are natural anti-oxidants that exhibit stronger T3 are found to be more promising than T for treatment of various
anti-cancer activity than tocopherol (T), the other analogues of cancers.
vitamin E [4].
4. Tocotrienols
2. Vitamin E
The natural vitamin E derivative, T3 was discovered in 1956. The
Vitamin E is a group of hydrophobic lipid-soluble compounds name “tocotrienol” was first proposed by Bunyan et al. to signify the
consisting of eight isoforms [8]. It was first discovered in 1922 as an isoprenoid side chain detected when it was isolated from the latex
essential fertility factor. Since its discovery, it has been considered of the rubber plant, Hevea brasiliensis. The anti-cancer property of
as one of the most potent anti-oxidants, neutralizing free radicals by T3 was identified and studied since 1985 [13,18]. The four isomers
donating hydrogen from its chromanol ring [9]. Vitamin E is abun- of T3 are distinguished based on the number and position of methyl
dantly found in edible oils including oils of barley, coconut, rice groups on the chromanol rings i.e. ␣-T3 is 5, 7, 8, trimethyl; ␤-T3
bran, palm, wheat etc. [10]. Vitamin E is primarily grouped into two is 5, 8, dimethyl; ␥-T3 is 7, 8, dimethyl and ␦-T3 is 8, monomethyl
categories namely, tocopherols (T) and tocotrienols (T3), each con- (Fig. 1) [10]. The T3 isoforms are mostly found at high concentra-
sisting of four isoforms namely: ␣-, ␤-, ␥- and ␦- (Fig. 1) [8,10,11]. tion in palm oil, rice bran oil and cereal grains including barley,
Tocopherols (T) are the saturated isoforms of vitamin E whereas oats, wheat, etc. [4]. Pharmacokinetics studies of T3 in rats sug-
tocotrienols (T3) are the unsaturated isoforms with isoprenoid side gested that oral administration provide the highest bioavailability
chain [10]. rate although the absorption was incomplete, whereas intramus-
cular and intraperitoneal routes of administration have negligible
3. Tocotrienols are superior to tocopherols absorption. However, among the isoforms, ␣-T3 has the highest
bioavailability, showing absolute bioavailability 28% while ␥-T3
Over past several decades, majority of vitamin E research was and ␦-T3 has 9%. A phase I enzyme cytochrome p450 3A4 (CYP3A4)
focused on ␣-tocopherol (␣T) isoform, as it was considered as the and P-glycoprotein of the gastrointestinal epithelium are associ-
most potent anti-oxidant isoform whereas only about 3% of the ated with oral absorption of T3 [10,19]. In addition, the unsaturated
study has been carried out for the T3 isoforms [9,12]. However, isoprenoid side chain of T3 contributes to their enhanced absorp-
accumulating preclinical and clinical studies have significantly tion into fatty tissues of the brain and liver and their distribution
proven that T3 display stronger anti-oxidant, anti-inflammatory, on the plasma membranes [9]. Further, the biological half-life (t1/2 )
and chemopreventive activities when compared with T [9,13]. The of ␣-T3, ␥-T3, and ␦-T3 in humans was determined as 4.4, 4.3,
B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272 261

Fig. 1. Structures of different analogues of Vitamin E. The figure has been made according to Roskoski et al. [188].

and 2.3 h, respectively [19]. Accumulated findings have strongly phoma 2 (Bcl-2), B-cell lymphoma-extra large (Bcl-xL), cellular
established T3 as analgesic, anti-ageing, anti-bacterial, anti- FLICE-like inhibitory protein (cFLIP), X-linked inhibitor of apopto-
cancer, anti-cholesterolemic, anti-diabetic, anti-hyperlipidemic, sis (XIAP), survivin etc. [3,34]. Moreover, T3 inhibited Src kinase,
anti-inflammatory, anti-oxidant, anti-pyretic, anti-thrombotic, Janus kinase (JAK) 1, and JAK2, which are the regulators of STAT3,
cardioprotective, chemopreventive, gastroprotective, hepatopro- thus eventually downregulating STAT3 (Fig. 2) [3,35]. Notably, T3
tective, and neuroprotective agents and are found to improve inhibited cancer cell proliferation by downregulating 3-hydroxy-3-
cognition, bone density etc. [4,10,13,20,21]. The anti-oxidant activ- methylglutaryl coenzyme A reductase (HMGCR), cyclin-dependent
ity of T3 is facilitated by activation of anti-oxidant enzymes such kinases (CDK)-2, −4, −6, c-Jun, c-Myc, mitogen-activated protein
as superoxide dismutase (SOD), NADPH: quinone oxidoreductase kinases (MAPK), phosphoinositide 3-kinase (PI3K)/Akt, Wnt/␤-
(NQO) and glutathione peroxidase (GPX) that scavenge free radi- catenin etc. [10,33,36–41]. Alternatively, T3 were also reported to
cals. In addition, studies have shown that T3 isoforms exert their induce growth arrest and apoptosis by activating CDK inhibitors
anti-cancer effect in cancer cells of the breast, colon, liver, lung, skin, (p21, p27, p53), caspase-3, -8, -9, Bcl-2-associated X (Bax) and
stomach, pancreas, prostate etc. [10]. Among the four T3 isoforms, enhancing cleavage of Bid, Apaf-1, Fas, etc. [10,33]. Additionally,
␥- and ␦-T3 were found to exhibit stronger anti-cancer activity [13]. T3 were found to downregulate angiopoietin-1, fibroblast growth
The anti-cancer activity of T3 is mediated via induction of apopto- factor (FGF), metalloproteinase-9 (MMP-9), TNF-␣, and vascular
sis, modulation of cell cycle progression, inhibition of angiogenesis, endothelial growth factor (VEGF), thereby inhibiting angiogenesis
metastasis etc. [22–24]. Moreover, combination of T3 with several in cancer [10,33,42].
chemotherapeutic drugs such as celecoxib, gefitinib, gemcitabine,
statins, or dietary components such as curcumin, polyphenols, 6. Tocotrienols as chemopreventive and chemotherapeutic
sesamin, etc. effectively sensitized the cancer cells to these agents agents for various cancers
[25]. Above all, several clinical trials have proven the efficacy, safety
and tolerability of T3 [26–28]. Kato et al. first reported the anti-cancer potential of T3 in 1985,
where ␣-T3 was shown to increase the life span of tumor-bearing
5. Molecular targets of tocotrienols in cancer rats [18]. Subsequently, numerous investigations have evinced the
anti-cancer potential of T3 in cancers of the breast, colorectal,
Aforementioned, T3 exert several inherent anti-cancer activi- liver, lung, pancreas, prostate, stomach etc. The next section of this
ties including anti-inflammatory, anti-proliferative, anti-survival, review will focus on the various preclinical and clinical findings of
anti-angiogenic properties etc., which are mediated via the reg- T3 against different cancers (Tables 1–3).
ulation of multiple signaling pathways associated with cancer
[29–32]. For instance, T3 were found to exert anti-inflammatory 6.1. Breast cancer
activity by suppressing COX-2, hypoxia-inducible factor-1 (HIF-1),
inducible nitric oxide synthase (iNOS), prostaglandin E2 (PGE2), Observational studies have suggested that vitamin E from
and interleukin (IL)-1,-6 and -8 [10,33]. Increasing lines of evidence dietary sources may prevent breast cancer in women [178]. Sup-
has ascertained that the anti-cancer activity of T3 was primarily porting this fact, accumulating evidence has substantiated that T3
mediated by the inhibition of two important transcription fac- exhibited chemopreventive and anti-cancer activity against breast
tors, NF-␬B and STAT3 and their regulated gene products [3]. For cancer both in vitro and in vivo (Fig. 2). For instance, T3 has been
example, T3 was shown to inhibit the tumor necrosis factor-alpha shown to reduce the growth of both estrogen-receptor-positive
(TNF-␣)-induced NF-␬B activation, which resulted in the downreg- (ER+) and estrogen-receptor-negative (ER-) phenotypes of breast
ulation of its related gene products that are involved in cell survival cancer cells by inhibiting HMGCR activity [48,179–181]. In addi-
including inhibitors of apoptosis proteins (IAP)-1, −2, B-cell lym- tion, it has also been proven that ␣-, ␥- and ␦-T3 suppressed the
262 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

Table 1
Tocotrienols Against Different Cancers.

Cancer In vitro/ in vivo Cell line/ Model Mechanism of action Reference

Bladder Cancer In vitro T24, 5637; J82; UMUC-3 ↓STAT3 [43]


Breast Cancer In vitroc T47D ↓Exosome heregulin [44]
Breast Cancer In vitroc MCF-7/Adr ↓P-gp [45]
Breast Cancer In vitroc MDA-MB-231; T-47D; MCF-10A ↓Wnt/␤-catenin [41]
Breast Cancer In vitroc SKBR3; BT474 ↓HER2 [46]
Breast Cancer In vitroc +SA ↓Rac1/WAVE2 [47]
Breast Cancer In vitro MCF-7; T47D; MDA-MB-23 ↓HMGR [48]
Breast Cancer In vitroc MCF-7; MDA-MB-231 ↑ER stress [6]
Breast Cancer In vitro MDA-MB-231; MDA-MB-468 ↑miRNA-429 [49]
Breast Cancer In vitro MCF-7 ↓RAS/ERK [50]
Breast Cancer In vitro +SA; MCF-7 ↓c-Myc [51]
Breast Cancer In vitroc +SA; MCF-7 ↓Akt/mTOR; ↓c-Myc [52]
Breast Cancer In vitroc +SA; MCF-7; MDAMB-231 ↓Akt/mTOR [53]
Breast Cancer In vivoi +SAj ↓Akt/mTOR [54]
Breast Cancer In vitro MDA-MB-231; MCF-7 ↓NF-␬B [55]
Breast Cancer In vitro SKBR3 ↑ROS [56]
Breast Cancer In vivo HER-2/neuk ↑HER-2/neu [56]
Breast Cancer In vitro – ↓PI3K/Akt [57]
Breast Cancer In vitrod SKBR3 ↓p38; ↓ERK1/2 [58]
Breast Cancer In vitrod,e 4T1 ↓IL-8; ↓VEGF [42]
Breast Cancer In vitroc MCF-7; MDA-MB 231 ↑ER stress [59]
Breast Cancer In vitroc – ↑JNK/CHOP/DR5 [60]
Breast Cancer In vitroc +SA ↓HGF; ↑Met [61]
Breast Cancer In vitro MCF-7 ↓AP15; ↑MIG6 [62]
Breast Cancer In vitroc,d SKBR3; MCF-7 ↑p53; ↑p21; ↑p16 [63]
Breast Cancer In vitroc MCF-7 ↑NQO2 [64]
Breast Cancer In vitroe MCF-7 ↑ER-␤ [65]
Breast Cancer In vitroc MCF-7; MDA-MB-231 ↓Id1 [66]
Breast Cancer In vitroc +SA ↓G1-S progression [30]
Breast Cancer In vitroa,c,d MDA-MB-231; MCF-7 ↑NRF2; ↓KEAP1 [67]
Breast Cancer In vitro MDA-MB-231; MCF-7 ↑JNK; ↑p38 MAPK; ↑DR5 [68]
Breast Cancer In vitrod,e 4T1 ↑IL-24; ↓VEGF; ↓IL-8 [69]
Breast Cancer In vivo 4T1j ↑IL-24 [69]
Breast Cancer In vitro MDA-MB-231 ↑ER-␤ [70]
Breast Cancer In vitro +SA ↑ER stress [71]
Breast Cancer In vivoe 4T1j ↓VEGF [72]
Breast Cancer In vitrod MDA-MB-231 ↓Cyclin D1; ↓pRb [73]
Breast Cancer In vitroc MCF-7 ↓NF-␬B [34]
Breast Cancer In vitroc +SA ↓PI3K/Akt [74]
Breast Cancer In vitroc – ↓Pl3K/PDK-1/ Akt [75]
Breast Cancer In vitro +SA ↓PI3K/PDK/Akt [76]
Breast Cancer In vitroc +SA ↑Bcl-2; ↑Bcl-xL [77]
Breast Cancer In vitroc +SA ↓Akt; ↓NF-␬B [78]
Breast Cancer In vitro MCF-7; MDA-MB 231 ↑MM1; ↑IFITM-1 [79]
Breast Cancer In vivo MCF-7l ↓c-Myc [80]
Breast Cancer In vitroc MDA-MB-231 ↑Mitochondrial disruption [81]
Breast Cancer In vitro +SA ↑Caspase-8 [82]
Breast Cancer In vitrod MDA-MB-435 ↑TGF- ␤ receptor II; ↑TGF-b, Fas, JNK [83]
Breast Cancer In vivoe DMBAm ↓HMG-CoA [84]
Breast Cancer In vitro CL-S1; -SA; +SA ↑DNA fragmentation [16]
Breast Cancer In vitro MCF-7; MDA-MB 435 ↑DNA fragmentation [85]
Cervical Cancer In vitroc HeLa ↑G0 /G1 cell cycle arrest [86]
Cervical Cancer In vitroc,d HeLa ↑ER stress [87]
Cervical Cancer In vitro HeLa ↑IL-6; ↓Cyclin D3; ↓p16; ↓CDK6 [31]
Colorectal Cancer In vivoc HCT 116n ↓NF-␬B [88]
Colorectal Cancer In vivoe SW620n ↓Wnt [38]
Colorectal Cancer In vitrod DLD-1 ↑Caspases; ↓pAkt [89]
Colorectal Cancer In vivof DLD-1l ↑p21; ↑p27; ↑Caspase-3, -9; ↓pAkt [89]
Colorectal Cancer In vitroc SW620 ↓Wnt [39]
Colorectal Cancer In vitro SW620 ↓Wnt [40]
Colorectal Cancer In vitroc HT-29 ↓␤-catenin/Tcf [90]
Colorectal Cancer In vitroc SW620 ↓Wnt [91]
Colorectal Cancer In vitroc HT-29 ↑Bax/ Bcl-2 ratio [92]
Colorectal Cancer In vitrod DLD-1 ↓HIF-1␣ [93]
Colorectal Cancer In vitrod DLD-1 ↓PI3K/PDK/Akt [94]
Colorectal Cancer In vivod DLD-1l ↓PI3K/PDK/Akt [94]
Colorectal Cancer In vitro DLD-1 ↓Telomerase activity [95]
Colorectal Cancer In vitroe RKO ↑p53 [96]
Gastric Cancer In vitroc SGC-7901 ↓ERK1/2 [97]
Gastric Cancer In vitro SGC-7901 ↓MMP-2, -9 [98]
Gastric Cancer In vitro SGC-7901 ↓␤-catenin [99]
Gastric Cancer In vitroc SGC-7901 ↑Caspase-3, -9 [100]
Gastric Cancer In vitro SGC-7901 ↓MAPK [101]
Glioblastoma In vitrob U87MG ↑Caspase-8 [102]
B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272 263

Table 1 (Continued)

Cancer In vitro/ in vivo Cell line/ Model Mechanism of action Reference

Glioblastoma In vitro U87MG ↑Caspase-8 [103]


Leukemia In vitroc,d CEM-SS ↑Nuclear fragmentation [12]
Leukemia In vitroc HL- 60; NB-4; Raji; SY-5Y ↑Caspase; ↑Cyt.c; ↑Bid cleavage [104]
Leukemia In vitroc KBM-5 ↓NF-␬B [34]
Liver Cancer In vitroc HepG2 ↑Peroxiredoxin-4 [105]
Liver Cancer In vivo HCCLM3 Luc2o ↓Akt/ mTOR [106]
Liver Cancer In vitroc HepG2; C3A; SNU-38; PLC/PRF5 ↓STAT3 [107]
Liver Cancer In vitro HepG2 ↓TG; ↓VLDL secretion [108]
Liver Cancer In vitro MH134 ↑ PARP fragmentation [109]
Liver Cancer In vivo MH134p ↑PARP fragmentation [109]
Liver Cancer In vitro Hep3B ↑Caspase-8, -9 [110]
Liver Cancer In vitrod HepG2 ↑S phase arrest [111]
Liver Cancer In vivo 4NQOq ↑S phase arrest [111]
Liver Cancer In vitro BNL 1ME A.7R.1 ↑Caspase 3; ↑DNA fragmentation [112]
Liver Cancer In vitro dRLh-84 ↑Caspase-3 [113]
Liver Cancer In vivo DEN & AAFr ↓GSH-Px; ↓GST; ↓GSSG-Rx [114]
Liver Cancer In vivo AAFr ↓GGT; ↓UDP-GT [115]
Lung Cancer In vitrob A549 ↑Caspase-8 [102]
Lung Cancer In vitro - ↑Caspase-8 [103]
Lung Cancer In vitrod A549; H1650 ↓Notch-1 [116]
Lung Cancer In vitro A549 ↓Akt; ↓HIF-2␣ [117]
Lung Cancer In vitroc H1299 ↓NF-␬B [34]
Lung Cancer In vitrog A549 ↓Ras & RhoA prenylation [118]
Mesothelioma In vitro H2452 ↓Ras [119]
Mesothelioma In vitro H2452 ↓HIF-2␣ [120]
Mesothelioma In vitrog H28 ↓STAT [121]
Multiple myeloma In vitroc U266; MM.1R, MM.1S ↓STAT3 [35]
Multiple myeloma In vitroc U266 ↓NF-␬B [34]
Neuroblastoma In vitroc SH-SY5Y ↑Caspase-9 [122]
Oral Cancer In vitroc SCC-4 ↓NF-␬B [34]
Pancreatic Cancer In vitrod L3.6pl; MIAPaCa-2 ↓VEGF; ↓MMP-9 [123]
Pancreatic Cancer In vivod L3.6plo ↓MMP-9 [123]
Pancreatic Cancer In vitrod MIA PaCa-2 ↑Bax [124]
Pancreatic Cancer In vitrod MIA PaCa-2; SW1990; BxPC-3 ↑p27(Kip1); ↓RAS-MEK-ERK [125]
Pancreatic Cancer In vivod MIA PaCa-2n ↑p27(Kip1); ↓pMAPK [125]
Pancreatic Cancer In vivo LSL-Kras(G12D/+); Pdx-1-Cres ↓MEK/ERK; ↓Akt; ↓NF-kB [126]
Pancreatic Cancer In vitro Panc-1; MIA PaCa-2; BxPC-3 ↓ErbB2 [36]
Pancreatic Cancer In vitroc BxPC-3; MIA PaCa-2; Panc-1 ↓NF-␬B [127]
Pancreatic Cancer In vivo MIA PaCa-2o ↓NF-␬B [127]
Pancreatic Cancer In vitroc MIA PaCa-2; PC3 ↓STAT3 [35]
Pancreatic Cancer In vivoc MIA PaCa-2o ↓STAT3 [35]
Pancreatic Cancer In vitro PANC-1; MIA PaCa-2; BxPC-3 ↓HMGR [128]
Prostate Cancer In vitro PC3 ↓Fyn/HIF-1␣ [129]
Prostate Cancer In vitro VcaP ↑p21; ↑p27 [130]
Prostate Cancer In vivo VCaPn ↑p21; ↑ p27 [130]
Prostate Cancer In vitro PC-3 ↓Src; ↓STAT3 [131]
Prostate Cancer In vitro PC-3; LNCaP ↑DHS; ↑DHC [132]
Prostate Cancer In vivo LNCaPn ↑DHS; ↑DHC [132]
Prostate Cancer In vitro CHO-7; SRD-15 ↓SREBP-2 [133]
Prostate Cancer In vitro PC-3; CRL-1435; LNCaP; CRL-1740 ↓TGF␤2 [134]
Prostate Cancer In vitro PC-3; DU145 ↓CD133/CD44 [135]
Prostate Cancer In vitrod PCa ↓NF-␬B; ↓EGFR [136]
Prostate Cancer In vitroe LNCaP; DU145; PC-3 ↑G0/ G1 cell cycle arrest [137]
Skin Cancer In vitrod BLM; A375 ↑ER-stress [138]
Skin Cancer In vivod A375n ↑ER-stress [138]
Skin Cancer In vitro B16 ↑ERK [139]
Skin Cancer In vitro B16F10 ↑Syntaxin7; ↑Vps16; ↑Vps3; ↑Vps41 [140]
Skin Cancer In vitrod A2058; A375 ↑Caspase-3; ↓CDK-4 [141]
Skin Cancer In vitro C32; G361 ↓NF-␬B; ↑JNK [142]

Note: ↑, Upregulation; ↓- Downregulation; AAF - 2-acetylaminofluorene; AP15- Apoptosis inhibitor 5 protein; Bax- BCL2-Associated X; Bcl-2 - B-cell lymphoma 2; Bcl-xL-
B-cell lymphoma-extra large; cAMP-Cyclic adenosine monophosphate; CDK-cyclin-dependent kinase; CDK6- Cyclin-dependent kinase 6; CHOP-CCAAT-enhancer-binding
protein homologous protein; Cyt.c - Cytochrome c; DEN -Diethylnitrosamine; DHC- Dihydroceramide; DHS- Dihydrosphingosine; DMBA- 7,12 Dimethylbenz(a)anthracene;
DR- death-receptor; EGFR-epidermal growth factor receptor; EGR-1-Early growth response protein 1; ER- Endoplasmic reticulum; ER-␤ -Estrogen receptor beta; ErbB-
erb-b2 receptor tyrosine kinase; ERK- extracellular signal–regulated kinase; GGT-gamma-glutamyltranspeptidase; GSSG-Rx-glutathione reductase; GSH-Px-glutathione per-
oxidase; GST- glutathione S-transferase; HER2-Human epidermal growth factor receptor 2; HGF- Hepatocyte growth factor; HIF-Hypoxia-inducible factor; HMGR- HMG-CoA
reductase; ID- inhibitor of DNA binding; ID1, Inhibitor of DNA binding 1; IFITM-1- interferon-inducible protein 9–27; IL-Interleukin; JNK - Jun amino-terminal kinases;
KEAP- Kelch Like ECH Associated Protein; MAPK-Mitogen-activated protein kinases; MIG6-Mitogen-inducible gene-6; MM1- c-Myc binding protein; MMP- Matrix metal-
loproteinases; mTOR- mechanistic target of rapamycin; NF-␬B- nuclear factor kappa-light-chain-enhancer of activated B cells; NQO2-NAD(P)H dehydrogenase quinone 2;
pAkt- phosphorylated Akt; PARP- Poly (ADP-ribose) polymerase; PDK- 3-phosphoinositide dependent protein kinase; PI3K, Phosphatidylinositol-4,5-bisphosphate 3-kinase;
P-gp- P-glycoprotein; Ras: rat sarcoma virus; pRb- phosphorylated retinoblastoma protein; RBT3- Rice bran tocotrienol; ROS- Reactive Oxygen Species; SREBP-2 -sterol-
regulatory-element-binding protein isoform 2; STAT3- Signal transducer and activator of transcription 3; T3-Tocotrienol; T3E- 6-O-carboxypropyl-alpha-tocotrienol; Tf-
Transferrin; TGF-␤ -Transforming growth factor beta; TG-triglyceride; TRF, Tocotrienol Rich Fraction; UDP-glucuronyltransferase; VEGF-Vascular endothelial growth factor;
VPS- Vacuolar protein sorting-associated protein; WAVE2, Wiskott–Aldrich Syndrome protein-family verprolin-homologous protein 2; XIAP- X-linked inhibitor of apoptosis
protein.
a
␣- T3; b ␤-T3; c ␥-T3; d ␦-T3; e TRF; f RBT3; g T3E; h TRF entrapped ␣-T3; i Oxazine derivatives of ␥- & ␦-T3; j BALB/c mice; k transgenic mice; l athymic nude mice;
m
Sprague–Dawley rats; n xenografts mice; o Orthotopic mice; p C3H/HeN mice; q ddY mice; r Rattus norvegicus; s Mutant mice.
264 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

Fig. 2. Tocotrienols can modulate different molecular targets in various cancers. The figure has been made according to Roskoski et al. [188].

proliferation of breast cancer cells by altering the expression of Apart from the T3 isoforms, other formulations of T3 also
oxidative stress modulatory enzymes GPX, SOD, NQO1 and NQO2, exert anti-tumor effects on breast cancer. For instance, tocotrienol
and cell cycle regulatory proteins (Rb/E2F complex, cyclin D1/CDK- oxazine derivatives inhibited mammary tumor growth by reduc-
4 and cyclin B1/CDK-1) [64,67]. The anti-proliferative effects of T3 ing pAkt, NF-␬B, COX-2, cyclin D1, CDK-2, −4, −6 and increasing
in mammary cancer cells are also associated with suppression of cell-cycle arrest proteins (p21 and p27) [54]. Likewise, another
c-Myc; inhibition in early post-receptor steps involved in cAMP form of T3, annatto-T3 that comprises of 90% ␦-T3 and 10% ␥-
production upstream from EGF-dependent MAPK and PI3K/Akt T3, reportedly delayed the development of mammary tumor and
signaling; suppression of Wnt/␤-catenin pathway; inhibition of reduced the number and size of the tumor via enhancing both apo-
Rac1/WAVE2 signaling; decreased AP15 and increased MIG6 genes ptosis and senescent-like growth arrest in HER-2/neu transgenic
and miRNA-429 [41,47,49,51,57,62,74,75,77,182]. Additionally, ␥- mice [5]. Furthermore, treatment of MCF-7-injected athymic mice
T3 also exerted its anti-proliferative activity by lipid raft disruption with tocotrienol-rich fraction (TRF) significantly downregulated c-
as well as decrease in exosome heregulin content, a potent lig- Myc and CD59 glycoprotein precursor gene involved in the immune
and for HER3 and HER4 receptors [44,46]. Interestingly, in vitro system, thus contributing to its anti-tumor effect [80]. Moreover,
treatment of mammary tumor cells with ␥-T3 resulted in reduced tocomin, which consists of a mixture of naturally occurring T3
glycolysis and pAkt/mechanistic target of rapamycin (mTOR) path- and T, induced apoptosis in breast cancer cells [144]. Hence, these
way [52]. Additionally, ␥-T3 also inhibited the mammary cancer investigations collectively indicate the possible benefits of T3 in
cell growth by inhibiting K-Ras, H-Ras, and pERK expression and prevention and treatment of breast cancer.
upregulating TGF␤-, Fas- and JNK-signaling pathway [50,82,83].
Moreover, in ER␤-containing breast cancer cells, T3 activated 6.2. Colorectal cancer
the ER␤ signaling pathway and enhanced the estrogen-responsive
genes such as MIC-1, subsequently conferring caspase-dependent Research over the past several years has revealed the potential
apoptosis [70]. Studies have also shown that T3 induce apopto- benefits of T3 for colorectal cancer. Studies have ascertained that
sis in breast cancer cells by increasing endoplasmic reticulum (ER) ␦-T3 significantly inhibit the growth of colon cancer cells through
stress and autophagy [6,59,68,71,144]. In addition to this, T3 also the downregulation of Wnt-1, ␤-catenin, c-jun, and cyclin D1 [40].
prompted apoptosis in vitro by inhibiting NF-␬B, PI3K/Akt/mTOR Similarly, ␥-T3 was also found to impede the proliferation of HT-
and subsequent downregulation of FLIP, increase of Bax/Bcl-2 ratio 29 colon cancer cells via downregulating Bcl-2 and upregulating
and PARP cleavage [55,57,75,183]. Another study revealed that the Bax and caspase-3 [92]. ␦-T3 was reported to reduce telomerase
apoptotic property of ␥-T3 in breast cancer cells is mediated by de activity by suppressing PKC activity in colorectal adenocarcinoma
novo ceramide synthesis dependent activation of JNK/CHOP/DR5 cells, which led to the downregulation of c-Myc and human tel-
signaling [60]. The anti-angiogenic effect of ␦-T3 against breast omerase reverse transcriptase (hTERT) expression, thus signifying
cancer was also mediated by its ability to reduce the synthesis its anti-proliferative role [95]. In addition to this, ␥-T3 also signif-
of proangiogenic markers such as IL-8 and VEGF [42,58,69,72,81]. icantly induced apoptosis in vitro and in vivo by downregulating
Besides these in vitro studies, several in vivo findings have also NF-␬B and its regulated gene products [88,92]. Besides, many
confirmed the anti-cancer activities of T3 against breast cancer. other studies have also reported that T3 repressed colon tumor
For instance, it has been successfully proven that T3 increased growth and induced apoptosis by suppressing pAkt, upregulating
tumor latency and reduced the tumor incidence and number in CDK inhibitors (p21, p27), caspase-3, and −9, and deregulating
7,12 dimethylbenz(a)anthracene-induced mammary tumor in rats ␤-catenin/Tcf (T-cell factor) signaling [89,90]. Notably, TRF also
[3]. induced apoptosis in colon cancer cells by activation of p53 and
B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272 265

alteration of Bax/Bcl2 ratio [96]. Moreover, an in vivo study has [10,107,109–111]. The relative anti-proliferative effect of T3 iso-
proven that TRF reduced the growth of human colon cancer mice forms against HCC cells were reported as ␦-T3 > ␤-T3 > ␣-T3 > ␥-T3
xenografts via suppression of Wnt pathway [38]. Further, studies [3,111]. ␥-T3 was shown to inhibit the proliferation of HCC cells
have also demonstrated that ␦-T3 hindered hypoxia-induced VEGF by upregulating peroxiredoxin-4 (Prx4) [105]. Additionally, ␥-T3
and IL-8 overexpression and by lowering HIF-1␣, thus consequently displayed its anti-proliferative and apoptotic functions in HCC
inhibiting angiogenesis in breast cancer cells [93,94]. Collectively, by upregulation of Bax, caspase-8 and caspase-9; increasing Bid
these studies indicate T3 as promising candidates for the treatment fragmentation; inhibiting STAT3 and its regulated gene prod-
of colon cancer (Fig. 2). ucts including Bcl-2, Bcl-xL, survivin, cyclin D1, Mcl-1, and VEGF
[107,110]. Besides, administration of T3 remarkably attenuated
6.3. Gastric cancer hepatocarcinogenesis induced by diethylnitrosamine (DEN)/2-
acetylaminofluorene (AAF) in rats [114,115]. Administration of
Increasing lines of evidence has confirmed the potential of T3 for ␥-T3 and ␦-T3 also remarkably inhibited the development of HCC in
the prevention and treatment of gastric cancer (Fig. 2). For instance, mouse xenograft and orthotopic models by abrogating Akt/mTOR
␥-T3 was reported to impede the growth of human gastric cancer signaling pathway [106,109]. Interestingly, both ␥-T3 and ␦-T3
in vitro and in vivo by inhibiting NF-␬B pathway [165]. Similarly, were detected only in tumor tissues, but not in normal tissues,
␥-T3 also induced apoptosis in gastric cancer cells by downregulat- signifying their distinct specificity to the tumor [109]. Addition-
ing Raf-ERK signaling pathway and activating caspase-9 [100,101]. ally, ␥-T3 also possesses lipid-lowering function in HCC, which may
Furthermore, it has been demonstrated that ␥-T3 inhibited HIF-1␣ in part contribute to its anti-cancer activity [108,186]. This lipid-
accumulation and paracrine secretion of VEGF through suppres- lowering activity is supported by an in vivo study, wherein ␥-T3
sion of pERK1/2 thus, substantially displaying its chemopreventive reduced triglyceride (TG) via regulating various lipogenic enzymes
and anti-angiogenic abilities [97]. It has also been revealed that ␥- such as fatty acid synthase (FAS), sterol regulatory element-binding
T3 downregulated MMP-2, −9 and upregulated tissue inhibitor of transcription factor 1(SREBF1), stearoyl CoA desaturase 1(SCD1)
metalloproteinase-1 (TIMP-1) and TIMP-2 in vitro, thus elucidating and carnitine palmitoyl transferase 1A (CPT-1A) [186]. Besides, T3
its anti-metastatic property against gastric cancer [98]. was also found to suppress the upstream regulators of lipid homeo-
stasis genes including diacylglycerol O-acyltransferase 2 (DGAT2),
6.4. Glioblastoma apolipoprotein B (ApoB)-100, SREBP1/2 and HMGCR, resulting in
the reduction of hepatic TGs, cholesterol and very low-density
In glioblastoma, the potential of ␦-T3 is elucidated by its ability lipoprotein (VLDL) secretion in HepG2 HCC cells [108]. Hence, these
to induce marked cytotoxicity in SF-295 glioblastoma cells in vitro reports collectively unveiled the potential of T3 in the prevention
(Fig. 2) [184]. More notably, all the isoforms of T3 have been shown and treatment of HCC.
to induce apoptosis in glioblastoma in vitro by activating caspase-8
and enhancing mitochondrial cytochrome c (cyt. c) release due to
loss of mitochondrial membrane integrity [102,103]. Likewise, T3
6.7. Lung cancer
formulation, TRF trapped in transferrin bearing vesicles was found
to remarkably enhance the cytotoxic activity of TRF by more than
In lung cancer, T3 isoforms may serve as the new efficacious
100 folds in glioblastoma T98G cells by improving the uptake of
alternatives for successful treatment (Fig. 2). In support of this,
TRF [185]. However, further in vitro and in vivo studies are vital to
Lim et al. revealed that all the T3 isoforms remarkably induced
determine the underlying mechanism of the anti-cancer effect of
apoptosis in lung cancer cells through activation of caspase-8 and
T3 in glioblastoma.
mitochondrial cyt.c release [102,103]. Further, ␦-T3 also exerted
its anti-cancer activity in lung cancer by elevating microRNA,
6.5. Leukemia
miR-34a, that led to the downregulation of Notch-1 and its
downstream targets such as Bcl-2, cyclin D1, and survivin [116].
In leukemia, only a few studies have elucidated the therapeu-
Interestingly, a redox-silent analogue of ␣-T3, 6-O-carboxypropyl-
tic action of T3. Yet, it has been found that the three isomers of
alpha-tocotrienol (T3E) was found to possess higher anti-cancer
T3 (␣-, ␥-, ␦-) exerted marked cytotoxic effect against human T-
potential than T3 in A549 lung cancer cells [117,118]. Additionally,
cell lymphoblastic leukemia cells [12]. Moreover, ␥-T3 imparted
studies showed that T3E suppressed hypoxia adaptation of lung
cytotoxicity in various leukemia cell types including lymphoblas-
cancer cells through the inhibition of Src-induced Akt activation
tic, myeloblastic and relapsed leukemia, portraying its potential
and decreased HIF-2␣ [117]. T3E was also reported to induce apo-
role in the treatment of leukemia [104]. Reports have indicated
ptosis in human lung adenocarcinoma that harbors Ras mutation
that ␦- and ␥-T3 induced apoptosis in leukemia cells in vitro via
[118].
nuclear chromatin condensation and nuclear fragmentation [12].
In addition, studies have revealed that ␥-T3 induced apoptosis in
leukemia cells by inhibiting NF-␬B, activating the caspase cascade,
increasing cyt.c release and Bid cleavage (Fig. 2) [34,104]. Further, 6.8. Mesothelioma
␥-T3 induced apoptosis in HL-60 leukemic cells by downregulating
two survival enzymes, HMGCR and glyoxalase 1 (GLO-1) via inhi- Malignant mesothelioma is an aggressive and highly chemore-
bition of Ras/Raf/ERK/NF-␬B and Ras/Akt/NF-␬B pathways [167]. sistant malignancy that requires effective treatment. It has been
Additionally, a mixture of ␦-T3 and its peroxy dimer was reported shown that T3E induced apoptosis in mesothelioma HT28 cell
to trigger apoptosis via mitochondrial pathway, thus reducing the through inhibition of Src and Src-independent STAT3 (Fig. 2)
growth and survival of leukemia cells [184]. [121]. Similarly, another study showed that T3E induced cyto-
toxicity in human mesothelioma H2452 cells by abrogating
6.6. Liver cancer cap-dependent translation complex formation through the inac-
tivation of oncogene Ras [119]. Moreover, T3E also detained
In recent years, several studies have confirmed the anti- cobalt (II) chloride-induced VEGF expression by downregulating
cancer effect of T3 against hepatocellular carcinoma (HCC) through HIF-2␣ signaling in mesothelioma cells thus, emphasizing its anti-
the deregulation of different key signaling pathways (Fig. 2) carcinogenic property [120].
266 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

6.9. Pancreatic cancer ␦-T3 was shown to induce G1 cell cycle arrest in A2058 and A375
human melanoma cells, via downregulating CDK-4 and activating
Several in vitro and in vivo studies have evinced the preventive caspase-3 [141]. Further, studies have shown that ␥-T3 induced
and therapeutic potential of T3 against pancreatic cancer (Fig. 2) apoptosis in melanoma cells via suppression of NF-␬B, EGFR, Id
[125]. For instance, ␥-T3 was found to remarkably suppresses pan- family proteins and JNK signaling pathway [142]. The anti-cancer
creatic cancer growth both in vitro and in vivo via inhibition of effect of ␦-T3 has also been elucidated by its ability to induce apo-
the constitutive NF-␬B activation and its regulated gene prod- ptosis via ER stress in melanoma both in vitro and in vivo [138].
ucts such as c-Myc, CXCR-4, cyclin D1, MMP-9, VEGF etc. [127]. Additionally, a recent in vivo study showed that intravenous admin-
Besides, ␦-T3 displayed its chemopreventive activity by inhibit- istration of ␣-T3 entrapped in transferrin-bearing vesicles caused
ing the progression of pancreatic intraepithelial neoplasm (PIN) in complete tumor eradication in 60% of B16-F10 melanoma tumor
Kras transgenic mouse model via inhibition of Kras-driven path- and extended the survival of mice [187]. Additionally, ␥-T3 treat-
ways such as MEK/ERK, PI3K/Akt and NF-␬B and enhanced Bax and ment halted cell invasion in human melanoma through inhibition
caspase-3 activities [126]. Further, in vitro as well as in vivo settings of mesenchymal markers and restoration of E-cadherin and ␥-
have confirmed that ␦-T3 induced G1 cell cycle arrest by elevating catenin [142]. Furthermore, ␦-T3 inhibited melanin formation in
nuclear accumulation of p27 (Kip1). Moreover, T3 also inhibited B16 melanoma cells through activation of ERK signaling pathway,
pancreatic cancer cell proliferation via suppression of ErbB2 and which in turn downregulated melanogenesis-related proteins such
mevalonate pathway, and activation of caspase-dependent pro- as microphthalmia-associated transcription factor (MITF), tyrosi-
grammed cell death [13,36,128]. Treatment of pancreatic cancer nase and tyrosinase-related proteins, TYRP-1 and TYRP-2 [139].
cells with ␦-T3 activated zinc finger transcription factor, EGR-1 Furthermore, it was also revealed that tocomin enhanced the
that enhanced the expression of Bax, consequently leading to apo- degradation of melanosomes in melanoma cells by upregulating
ptosis [124]. Further, a recent study has demonstrated that ␦-T3 endosome docking/fusion proteins including syntaxin7, vacuolar
significantly inhibited the metastasis of pancreatic ductal adeno- protein sorting-associated protein Vps16, Vps33, and Vps41 [140].
carcinoma (PDAC) and PDAC stem-like cells in vivo [123]. Collectively, these reports have shown the growing potential of T3
for skin cancer treatment.
6.10. Prostate cancer
6.12. Other cancers
In venture of novel drug discovery for prostate cancer, T3 was
found to have high potential in the prevention and treatment
Apart from the aforementioned cancers, T3 have high poten-
of prostate cancer (Fig. 2) [136]. In vitro studies showed that ␥-
tial in the prevention and treatment of many other cancers. For
T3 promoted apoptosis and autophagy in human prostate cancer
instance, studies showed that ␥-T3 act as a Bcl-2 homology domain
cells by elevating intracellular levels of sphingolipid intermediates
3 (BH3) mimetic in neuroblastoma SH-SY5Y cells by attaching to the
(dihydroceramide and dihydrosphingosine) involved in de novo
hydrophobic groove of Bcl-2 and promoted apoptosis via enhanc-
synthesis of spingolipids. Moreover, supplementation of ␥-T3 also
ing Bax and caspase-9 [122]. Another study indicated that ␥-T3
significantly reduced the tumor growth of LNCaP-xenograft mouse
abrogated NF-␬B activity in SCC-4 tongue squamous carcinoma
model [132]. Additionally, it has been shown that annatto-T3
cells thereby, enhancing apoptosis [34]. It was also found that ␦-T3
inhibited androgen-independent prostate cancer cell proliferation
induced apoptosis via inhibition of STAT3 pathway in human blad-
through simultaneous inhibition of tyrosine kinase, Src and STAT3
der cancer [43]. Furthermore, it has been reported that ␣- and ␥-T3
[131]. Further, studies have revealed that ␥-T3 induced apopto-
induced apoptosis in HeLa cervical cancer cells via enhancing Bax,
sis of prostate cancer cells by inhibiting TGF␤2, NF-␬B, EGFR,
cyt.c release, and IL-6, and downregulating Bcl-2, cyclin D3, CDK6,
Fyn/HIF-1␣ and Id family proteins (Id1 and Id3) and activating JNK-
and p16 [31,86]. Additionally, ␥- and ␦-T3 also induced apopto-
signaling pathway [129,134,136]. Interestingly, ␥-T3 executed its
sis in cervical cancer in vitro by promoting ER stress [87]. Further,
anti-invasive effect by suppressing mesenchymal markers and sig-
in multiple myeloma, ␥-T3 inhibited NF-␬B and STAT3 and their
nificantly improving the expression of E-cadherin and ␤-catenin
regulated gene products such as Bcl-2, cyclin D1 etc. thus, indicat-
[136]. It has also been reported that ␥-T3 substantially attenu-
ing its potential as an effective treatment alternative for multiple
ated the expression of prostate cancer stem cells (CSC) markers
myeloma patients (Fig. 2) [34,35].
CD133/CD44, which are predicted to be the main cause of remission
in androgen-independent prostate cancer [135]. Further, T3 was
found to degrade the sterol regulatory element-binding protein-2 7. Chemosensitizing effect of tocotrienols
(SREBP-2) in prostate cancer cells, thereby reducing their viability
through reduced cholesterol level [133]. Due to the development of chemoresistance in cancer cells, the
Likewise, TRF also portrayed its chemopreventive and therapeu- quest for finding novel and effective chemosensitizing agents has
tic activities against prostate cancer by inducing cell cycle arrest never ceased. Recent studies have affirmed the high potential of
and apoptosis in vitro [137]. Moreover, daily supplementation of T3 in chemosensitizing cancer cells to therapeutic agents through
TRF to transgenic adenocarcinoma mouse prostate mouse model modulation of various molecular targets (Table 2). For instance,
(TRAMP) significantly lowered the occurrence of tumor forma- in vitro and in vivo studies revealed that ␥-T3 sensitized pancreatic
tion and high-grade neoplastic lesions due to the upregulation of cancer cells to gemcitabine and inhibited pancreatic tumor growth
proapoptotic proteins Bcl-2 antagonist of cell death (BAD), caspase- by downregulating the NF-␬B-mediated inflammatory pathways
3 and CDK inhibitors (p21 and p27) [3,155]. linked to tumorigenesis [127]. Apart from gemcitabine, T3 also aug-
mented the chemosensitivity of pancreatic cancer cells to other
6.11. Skin cancer drugs such as ferulic acid, lovastatin etc. [128,145]. Synergistic
anti-cancer activities were also observed in prostate cancer upon
Since the past several decades, tocotrienols have been of great combined treatment with T3 and other agents including doc-
importance in dermatology for their protection against photosen- etaxel and lovastatin [169,175]. Congregate evidences have shown
sitivity. Recently, T3 have been proven effective against skin cancer that T3 in combination with therapeutic agents such as celecoxib,
(Fig. 2). For instance, ␦-T3 and its peroxy dimer were found to effec- curcumin, docosahexanoic acid, epigallocatechin gallate (EGCG),
tively impede the proliferation of melanoma cells [184]. Moreover, erlotinib, ferulic acid, gefitinib, oridonin, resveratrol, sesamin,
B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272 267

Table 2
Chemosensitizing action of tocotrienols.

Cancer Combination In vitro/ In vivo Mechanism of action Reference

Bladder Cancer ␦-T3 + GEM In vitro ↓STAT3 [43]


Breast Cancer ␦-T3 NE + CUR In vitro ↓NF-␬B [25]
Breast Cancer ␥-T3 + DHA In vitro ↓STAT3 [143]
Breast Cancer Tocomin In vitro ↓PI3K; ↓mTOR [144]
Breast Cancer ␥-T3 + oridonin In vitro ↓Akt/mTOR [7]
Breast Cancer ␦-T3 + ferulic acid In vitro ↑G1 cell cycle arrest [145]
Breast Cancer ␥-T3 + PPAR␥ antagonist In vitro ↓COX-2; ↓PGDS; ↓PGD2 [146]
Breast Cancer ␥-T3 + SU11274 In vitro ↓Akt; ↓STAT1/5; ↓NF-␬B [147]
Breast Cancer TRF + DC+TL In vivo ↑IFN-␥; ↑IL-12 [148]
Breast Cancer ␥-T3 + sesamin In vitro ↓cyclin D1; ↓CDK-2,-4,-6 [149]
Breast Cancer ␥-T3 + PPAR␥ antagonist In vitro ↓Akt [150]
Breast Cancer ␥-T3 + statin In vitro ↓HMGCR [151]
Breast Cancer ␥-T3 + SVA In vitro ↓STAT3 [152]
Breast Cancer ␥-T3 + sesamin In vitro ↓EGF [153]
Breast Cancer ␥-T3 + SU11274 In vitro ↓Met [154]
Breast Cancer Tocomin In vivo ↑BAD [155]
Breast Cancer ␥-T3 + CXB In vitro ↓Akt; ↓NF-␬B [156]
Breast Cancer ␥-T3 + erlotinib In vitro ↓ErbB3; ↓ErbB4 [157]
Breast Cancer ␥-T3 + gefitinib In vitro ↓ErbB3; ↓ErbB4 [157]
Breast Cancer ␥-T3 + gefitinib/ erlotinib In vitro ↓STAT; ↓Akt [158]
Breast Cancer ␥-T3 + statin In vitro ↑p27; ↓cyclin D1; ↓CDK2 [159]
Breast Cancer ␥-T3 + EGCG + resveratrol In vitro ↑NQO1 [160]
Breast Cancer ␥-T3 + statin In vitro ↓pMAPK; ↓JNK; ↓Akt [161]
Colorectal Cancer ␥-T3 + capecitabine In vivo ↓NF-␬B [88]
Colorectal Cancer ␦-T3 + jerantinine B In vitro ↓Caspase-8, -3 [162]
Colorectal Cancer ␥-T3 + ATST In vitro ↓HMGCR [163]
Colorectal Cancer ␥-T3 + ATST + CXB In vitro ↑G0 /G1 phase cell cycle arrest [164]
Colorectal Cancer ␥-T3 + TRAIL In vitro ↑DRs [35]
Gastric Cancer ␥-T3 + capecitabine In vitro ↓NF-␬B [165]
Gastric Cancer ␥-T3 + capecitabine In vivo ↓Ki-67; ↓CD31; ↓NF-␬B [165]
Glioblastoma ␦-T3 + jerantinine B In vitro ↑Caspase-3,-8 [162]
Glioblastoma ␥-T3 + HC In vitro ↑Caspase-3 [166]o
Leukemia ␥-T3 + LVT In vitro ↓NF-␬B [167]
Liver Cancer T3 + EPI-NPs In vitro ↓VEGF [168]
Liver Cancer T3 + EPI-NPs In vivo ↓VEGF [168]
Liver Cancer ␦-T3 + PTX In vitro ↓STAT3 [107]
Liver Cancer ␦-T3 + DOX In vitro ↓STAT3 [107]
Lung Cancer ␦-T3 + cisplatin In vitro ↓Notch-1 [116]
Lung Cancer ␦-T3 + LVT In vitro ↓HMGCR [169]
Mesothelioma TRF + cisplatin In vitro ↓Akt [170]
Mesothelioma ␦-T3 + statin In vitro ↓MVL pathway; ↑caspase-3; ↑ER stress [171]
Oral Cancer ␦-T3 + DTX In vitro ↓NF-␬B [172]
Ovarian Cancer ␦-T3 NE + CUR In vitro ↓NF-␬B [25]
Pancreatic Cancer ␦-T3 + ferulic acid In vitro ↑p21 [145]
Pancreatic Cancer ␦- T3 + GEM In vivo ↓VEGF; ↓pAkt [126]
Pancreatic Cancer ␦- T3 + GEM In vitro ↓NF-␬B [173]
Pancreatic Cancer ␦- T3 + GEM In vivo ↓NF-␬B [173]
Pancreatic Cancer ␦- T3 + GEM In vivo ↓NF-␬B [127]
Pancreatic Cancer d-␦-T3 + LVT In vitro ↓HMGR [128]
Prostate Cancer ␥-T3 + PSP In vitro ↑AMPK; ↓ACC [174]
Prostate Cancer ␥-T3 + DTX In vivo ↓Ki-67; ↓Id1 [175]
Prostate Cancer ␥-T3 + DTX In vitro ↑Caspases; ↓EGFR; ↓NF-␬B [136]
Prostate Cancer ␦-T3 + LVT In vitro ↓HMCGR [169]
Skin Cancer d-␦-T3 + LVT In vitro ↑Caspase-3; ↓HMCGR [141]
Skin Cancer ␥-T3+ ␦-T3+ LVT In vitro ↓HMCGR [169]

Note: ↑- Upregulation; ↓- Downregulation; ACC- acetyl-CoA carboxylase; ALDH- Aldehyde dehydrogenase; AMPK-AMP-activated protein kinase;ATST- atorvastatin; BAD Bcl-
2 antagonist of cell death; CD31- cluster of differentiation 31; COX-2- Cyclooxygenase2; CXB-Celecoxib; DC- dendritic cells; DHA- docosahexanoic acid; DOX-doxorubicin;
DR- death receptor; DTX-docetaxel; EGCG-Epigallocatechin gallate; EGF - Epidermal growth factor; EPI-Epirubicin; G1- Gap1; GEM-Gemcitabine; GLO1: Glyoxalase 1;
HC-hydroxychavicol; HMGCR-HMG-CoA- 3-hydroxy-3-methyl-glutaryl-CoA reductase; IFN␥- Interferon gamma; IL-12-Interleukin 12; JNK - Jun amino-terminal kinases;
LVT-Lovastatin; MAPK- Mitogen-activated protein kinases; MVL-mevalonate; mTOR- mechanistic target of rapamycin; NE-Nanoemulsion; NF-␬B- nuclear factor kappa-
light-chain-enhancer of activated B cells; NPs-Nanoparticles; NQO1- NAD(P)H dehydrogenase quinone 1; PGD2-prostaglandin D2; PGDS -prostaglandin synthase; PI3 K
-phosphoinositide 3-kinase; PPAR␥- Peroxisome proliferator-activated receptor gamma; PSP-Polysaccharopeptide; PTX- paclitaxel; RXR-retinoid X receptor; SCID- severe
combined immunodeficiency; STAT3-Signal transducer and activator of transcription 3; SVA -Simvastatin; T3- Tocotrienol; Tf -Transferrin; THD-thalidomide; TL- Tumor
lysate; Tocomin- Mixed tocotrineols with tocopherols; TPGS-d-␣-tocopheryl polyethylene glycol 1000 succinate; TRAIL-Tumor necrosis factor (TNF)-related apoptosis-
inducing ligand; TRAMP- transgenic adenocarcinoma mouse prostate; TRF- Tocotrienol rich fraction;VEGF- Vascular endothelial growth factor.

statin, etc., considerably sensitized breast cancer cells by down- antinine B, etc. [88,162,163] (Table 2). T3 also sensitized gastric
regulating Akt, NF-␬B, STAT3 and their associated gene products cancer cells to capecitabine both in vitro and in vivo through down-
(Table 2) [7,25,60,143,145,149,157,156,160]. Moreover, treatment regulation of NF-␬B-regulated markers of proliferation, invasion,
of bladder cancer cells with ␦-T3 and gemcitabine remarkably angiogenesis, and metastasis [165]. Combinatorial therapy com-
enhanced the potential of gemcitabine [50]. In colorectal can- prising of T3 either with jerantinine B or hydroxychavicol induced
cer, T3 exhibited chemosensitizing activity when combined with apoptosis of glioblastoma cells via enhancing caspase activation
other compounds such as atorvastatin, capecitabine, celecoxib, jer- [162,166]. Further, combination of T3 with chemotherapeutic
268 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

Table 3
Clinical Studies of Tocotrienols against Different Cancers.

Cancer Phase No. of pts Dose; duration Outcome/ status Reference

Breast Cancer Pilot trial 248 200 mg/daya ; 5yrs No significant outcome [176]
Breast Cancer II 78 900 mg/dayc Ongoing [177]
Colorectal Cancer II 70 900 mg/dayb Ongoing [177]
Lung Cancer III 190 900 mg/dayd Ongoing [177]
Ovarian Cancer II 23 900 mg/daye Ongoing [177]
Ovarian Cancer II 34 900 mg/day Ongoing [177]
Pancreatic Cancer I 25 200–1600 mg/day; 2 weeks Well tolerated; reached bioactive levels in blood [27]
a
Given in combination with tamoxifen.
b
Given in combination with FOLFOXIRI (5-fluorouracil, oxaliplatin, irinotecan).
c
Given in combination with neoadjuvant chemotherapy (epirubicin, cyclophosphamide, docetaxel, trastuzumab, pertuzumab).
d
Given in combination with cisplatin, vinorelbine, carboplatin.
e
Given in combination with bevacizumab.

agents such as paclitaxel, doxorubicin, and nano-encapsulated involving 190 patients with advanced non-small cell lung cancer
epirubicin enhanced apoptosis in liver cancer cells by downreg- is being carried out to evaluate the synergistic potential of ␦-T3
ulating VEGF and STAT3 [107,168]. Additionally, in lung cancer and in combination with standard chemotherapy. In addition, another
mesothelioma, it has been reported that T3 sensitized the can- phase II clinical trial is also being undertaken wherein advanced
cer cells to cisplatin and statin [116,169]. In vitro treatment of ovarian cancer patients are administered 300 mg × 3/day of T3 with
oral cancer cells with ␦-T3 and docetaxel significantly enhanced bevacizumab. Similarly, in another ongoing phase II clinical study,
the chemosensitivity by downregulating NF-␬B-regulated gene a daily dose 900 mg of T3 is orally administered to ovarian cancer
products concomitant with inhibition of apoptosis [172]. Further, patients until progression or unacceptable toxicity [177]. Comple-
combination of ␦-T3 and lovastatin synergistically induced apo- tion of these ongoing trials will provide us a solid foundation for
ptosis in human melanoma cells by inhibiting HMCGR activity further studies on the efficacy of tocotrienols for the management
[141] (Table 2). Hence, these preclinical findings have provided the of different cancers.
potential of T3 isoforms as novel chemosensitizing agents for the
effective treatment of cancer. 9. Conclusions and future prospect

8. Clinical studies on tocotrienols- completed and ongoing Preclinical studies conducted over the past several years have
established T3 as novel, safe, and efficacious multi-targeted anti-
Promising results from preclinical studies have encouraged few cancer agents. It has been well proven that T3 have higher efficacy
clinical studies where the efficacy of T3 against different cancers and health benefits when compared to T. Comprehensive studies
has been assessed. So far, only two clinical studies have been have substantiated the potential of T3 in modulating numerous key
completed, and five clinical trials are ongoing to evaluate the ther- signaling pathways involved in the development and progression
apeutic role of tocotrienols in breast, colorectal, lung and ovarian of cancer, thus encouraging their use as mono-targeted therapy as
cancers (Table 3). well as combinatorial therapy with other chemotherapeutic drugs.
The first clinical trial of T3 on cancer was published in 2010. Besides, several clinical studies have confirmed the safety and effi-
This study was carried out to assess the efficacy of T3 in combi- cacy of T3. However, potential of T3 are often constrained by their
nation with the drug, tamoxifen for breast cancer. In this study, poor solubility and low bioavailability. In recent years T3 are formu-
240 women, aged between 40 and 60 years with early breast lated in the form of nanoemulsions, liposomes, micelles complexes
cancer received TRF and tamoxifen for five years. However, the as well as lipid conjugates with chemotherapeutic drugs to enhance
results were not significant and showed no association between their bioavailability and efficacy. However, these formulations still
the adjuvant T3 therapy and breast cancer specific survival [176]. demand further investigations. Thus, in depth studies to find effi-
Subsequently, a phase I trial was conducted in 25 patients with cient delivery systems and increase the bioavailability of T3 are of
pancreatic ductal neoplasia. In this study, the patients received an prime importance [4,14]. Additionally, scarcity of T3 in nature and
escalating dose of 200–3200 mg of ␦-T3 for two weeks prior to their cost of extraction also increase their limitation, making T3
surgery and one dose on the day of surgery. It was found that a dose isoforms expensive compounds. Hence, further research for better
of 200–1600 mg of ␦-T3/daily for two weeks prior to surgery was extraction and synthesis methods of T3 isoforms is vital to attain
well tolerated, attained bioactive levels in the blood, and remark- their maximum value. Also, extensive investigation is warranted
ably instigated apoptosis of neoplastic cells, thus suggesting the to demarcate the different mechanisms of actions associated with
therapeutic potentials of ␦-T3 for pancreatic cancer [27]. In addi- different isoforms of T3 to establish them as alternatives in cancer
tion, several ongoing clinical studies of T3 are being carried out in therapeutic cache [17]. Furthermore, till today, only two clinical
Vejle hospital, Department of Oncology, Vejle, Denmark. For exam- trials of T3 on cancer patients have been completed, out of which
ple, a phase II clinical study is being conducted with breast cancer T3 was shown to have no significant outcome in breast cancer
patients to investigate the effect of T3 in combination with neoad- patients even though preclinical settings have revealed the incred-
juvant chemotherapy. In this study, a dose of 300 mg × 3/day of T3 is ible potential of T3 against breast cancer. Hence, further preclinical
given with chemotherapeutic agents such as epirubicin, docetaxel, and clinical studies are unquestionably indispensable in future to
cyclophosphamide, trastuzumab and ertuzumab. Further, another establish T3 as safe and promising agents, opening new avenues for
ongoing phase II study with metastatic colorectal cancer patients the prevention and treatment of different cancers.
aims to determine whether T3 can decrease the side effects of FOL-
FOXIRI (5-fluorouracil, oxaliplatin, irinotecan). In this particular Acknowledgement
trial, 70 colorectal adenocarcinoma patients are orally adminis-
tered with 900 mg/day of T3 in combination with FOLFOXIRI. In This work was supported by BT/529/NE/TBP/2013 awarded to
addition, the chemosensitizing effect of T3 has been carried out in Dr. Ajaikumar B. Kunnumakkara. The author Bethsebie Lalduhsaki
two ongoing breast cancer phase II trials. Further, a phase III trial Sailo acknowledges DST-INSPIRE for providing the fellowship. The
B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272 269

author, Kishore Banik acknowledges UGC, New Delhi for providing pharmacodynamic presurgical trial of vitamin E ␦-tocotrienol in patients
the fellowship. with pancreatic ductal neoplasia, EBioMedicine 2 (2015) 1987–1995.
[28] C.L. Shen, H. Mo, S. Yang, S. Wang, C.K. Felton, M.D. Tomison, I.N. Soelaiman,
Safety and efficacy of tocotrienol supplementation for bone health in
Conflict of interests postmenopausal women: protocol for a dose-response double-blinded
placebo-controlled randomised trial, BMJ Open 6 (2016) e012572.
[29] A. Kamal-Eldin, L.A. Appelqvist, The chemistry and antioxidant properties of
The authors declare that there are no conflict of interests asso- tocopherols and tocotrienols, Lipids 31 (1996) 671–701.
ciated with the manuscript. [30] G.V. Samant, V.B. Wali, P.W. Sylvester, Anti-proliferative effects of
gamma-tocotrienol on mammary tumour cells are associated with
suppression of cell cycle progression, Cell Prolif. 43 (2010) 77–83.
References [31] S.J. Wu, L.T. Ng, Tocotrienols inhibited growth and induced apoptosis in
human HeLa cells through the cell cycle signaling pathway, Integr. Cancer
[1] J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo, D.M. Ther. 9 (2010) 66–72.
Parkin, D. Forman, F. Bray, Cancer incidence and mortality worldwide: [32] T. Miyazawa, A. Shibata, K. Nakagawa, T. Tsuzuki, Anti-angiogenic function
sources, methods and major patterns in GLOBOCAN 2012, Int. J. Cancer 136 of tocotrienol, Asia Pac. J. Clin. Nutr. 17 (2008) 253–256.
(2015) E359–386. [33] A. Abdullah, A. Atia, Tocotrienols molecular aspects beyond its antioxidant
[2] W.P. Steward, K. Brown, Cancer chemoprevention: a rapidly evolving field, activity, J. Med. Res. Pract. N. Am. 2 (2013) 246–250.
Br. J. Cancer 109 (2013) 1–7. [34] K.S. Ahn, G. Sethi, K. Krishnan, B.B. Aggarwal, Gamma-tocotrienol inhibits
[3] M.T. Ling, S.U. Luk, F. Al-Ejeh, K.K. Khanna, Tocotrienol as a potential nuclear factor-kappaB signaling pathway through inhibition of
anticancer agent, Carcinogenesis 33 (2012) 233–239. receptor-interacting protein and TAK1 leading to suppression of
[4] M.M. Kanchi, M.K. Shanmugam, G. Rane, G. Sethi, A.P. Kumar, Tocotrienols: antiapoptotic gene products and potentiation of apoptosis, J. Biol. Chem. 282
the unsaturated sidekick shifting new paradigms in vitamin E therapeutics, (2007) 809–820.
Drug Discov. Today 22 (2017) 1765–1781. [35] R. Kannappan, S.C. Gupta, J.H. Kim, B.B. Aggarwal, Tocotrienols fight cancer
[5] M. Shen, T.H. Chan, Q.P. Dou, Targeting tumor ubiquitin-proteasome by targeting multiple cell signaling pathways, Genes Nutr. 7 (2012) 43–52.
pathway with polyphenols for chemosensitization, Anticancer Agents Med. [36] S. Shin-Kang, V.P. Ramsauer, J. Lightner, K. Chakraborty, W. Stone, S.
Chem. 12 (2012) 891–901. Campbell, S.A. Reddy, K. Krishnan, Tocotrienols inhibit AKT and ERK
[6] R.V. Tiwari, P. Parajuli, P.W. Sylvester, ␥-Tocotrienol-induced endoplasmic activation and suppress pancreatic cancer cell proliferation by suppressing
reticulum stress and autophagy act concurrently to promote breast cancer the ErbB2 pathway, Free Radic. Biol. Med. 51 (2011) 1164–1174.
cell death, Biochem. Cell Biol. 93 (2015) 306–320. [37] A. Shibata, K. Nakagawa, T. Tsuduki, T. Miyazawa, ␦-Tocotrienol treatment is
[7] R.V. Tiwari, P. Parajuli, P.W. Sylvester, Synergistic anticancer effects of more effective against hypoxic tumor cells than normoxic cells: potential
combined ␥-tocotrienol and oridonin treatment is associated with the implications for cancer therapy, J. Nutr. Biochem. 26 (2015) 832–840.
induction of autophagy, Mol. Cell. Biochem. 408 (2015) 123–137. [38] J.S. Zhang, S.J. Zhang, Q. Li, Y.H. Liu, N. He, J. Zhang, P.H. Zhou, M. Li, T. Guan,
[8] E. Cardenas, R. Ghosh, Vitamin E: a dark horse at the crossroad of cancer J.R. Liu, Tocotrienol-rich fraction (TRF) suppresses the growth of human
management, Biochem. Pharmacol. 86 (2013) 845–852. colon cancer xenografts in Balb/C nude mice by the Wnt pathway, PLoS One
[9] H.Y. Peh, W.S. Tan, W. Liao, W.S. Wong, Vitamin E therapy beyond cancer: 10 (2015) e0122175.
tocopherol versus tocotrienol, Pharmacol. Ther. 162 (2015) 152–169. [39] J.S. Zhang, D.M. Li, Y. Ma, N. He, Q. Gu, F.S. Wang, S.Q. Jiang, B.Q. Chen, J.R.
[10] B.B. Aggarwal, C. Sundaram, S. Prasad, R. Kannappan, Tocotrienols, the Liu, ␥-Tocotrienol induces paraptosis-like cell death in human colon
vitamin E of the 21 st century: its potential against cancer and other chronic carcinoma SW620 cells, PLoS One 8 (2013) e57779.
diseases, Biochem. Pharmacol. 80 (2010) 1613–1631. [40] M.A. Yue, N. He, J. Zhang, D. Li, Y. Liu, J. Zhang, Experimental study on
[11] C.K. Sen, S. Khanna, C. Rink, S. Roy, Tocotrienols: the emerging face of delta-tocotrienol inhibits the Wnt pathway in the colon cancer cell SW620,
natural vitamin E, Vitam. Horm. (2007) 203–261. Wei Sheng Yan Jiu 41 (2012) 900–904.
[12] R.S. Wong, A.K. Radhakrishnan, T.A. Ibrahim, S.K. Cheong, ␦- and [41] R.A. Ahmed, O.A. Alawin, P.W. Sylvester, ␥-Tocotrienol reversal of
␥-tocotrienols induce classical ultrastructural apoptotic changes in human T epithelial-to-mesenchymal transition in human breast cancer cells is
lymphoblastic leukemic cells, Microsc. Microanal. 18 (2012) 462–469. associated with inhibition of canonical Wnt signalling, Cell Prolif. 49 (2016)
[13] H. Ahsan, A. Ahad, J. Iqbal, W.A. Siddiqui, Pharmacological potential of 460–470.
tocotrienols: a review, Nutr. Metab. (Lond.) 11 (2014) 52. [42] K.R. Selvaduray, A.K. Radhakrishnan, M.K. Kutty, K. Nesaretnam, Palm
[14] A. Abu-Fayyad, M.M. Kamal, J.L. Carroll, A.M. Dragoi, R. Cody, J. Cardelli, S. tocotrienols decrease levels of pro-angiogenic markers in human umbilical
Nazzal, Development and in-vitro characterization of nanoemulsions loaded vein endothelial cells (HUVEC) and murine mammary cancer cells, Genes
with paclitaxel/␥-tocotrienol lipid conjugates, Int. J. Pharm. 536 (2017) Nutr. 7 (2012) 53–61.
146–157. [43] C. Ye, W. Zhao, M. Li, J. Zhuang, X. Yan, Q. Lu, C. Chang, X. Huang, J. Zhou, B.
[15] Q. Jiang, Natural forms of vitamin e as effective agents for cancer prevention Xie, Z. Zhang, X. Yao, J. Yan, H. Guo, ␦-tocotrienol induces human bladder
and therapy, Adv. Nutr. 8 (2017) 850–867. cancer cell growth arrest, apoptosis and chemosensitization through
[16] B.S. McIntyre, K.P. Briski, A. Gapor, P.W. Sylvester, Antiproliferative and inhibition of STAT3 pathway, PLoS One 10 (2015) e0122712.
apoptoticv effects of tocopherols and tocotrienols on preneoplastic and [44] O.A. Alawin, R.A. Ahmed, V. Dronamraju, K.P. Briski, P.W. Sylvester,
neoplastic mouse mammary epithelial cells, Proc. Soc. Exp. Biol. Med. 224 ␥-Tocotrienol-induced disruption of lipid rafts in human breast cancer cells
(2000) 292–301. is associated with a reduction in exosome heregulin content, J. Nutr.
[17] R. Comitato, R. Ambra, F. Virgili, Tocotrienols a family of molecules with Biochem. 48 (2017) 83–93.
specific biological activities, Antioxidants (Basel) 6 (2017) (pii: E93). [45] Y. Ding, Y. Peng, L. Deng, J. Fan, B. Huang, Gamma-tocotrienol reverses
[18] A. Kato, M. Yamaoka, A. Tanaka, K. Komiyama, I. Umezawa, Physiological multidrug resistance of breast cancer cells with a mechanism distinct from
effect of tocotrienol, J. Japan Oil Chem. Soc. (Yukugaku) 34 (1985) 375–376. that of atorvastatin, J. Steroid Biochem. Mol. Biol. 167 (2017) 67–77.
[19] S.P. Yap, K.H. Yuen, A.B. Lim, Influence of route of administration on the [46] O.A. Alawin, R.A. Ahmed, B.A. Ibrahim, K.P. Briski, P.W. Sylvester,
absorption and disposition of alpha-: gamma- and delta-tocotrienols in rats, Antiproliferative effects of ␥-tocotrienol are associated with lipid raft
J. Pharm. Pharmacol. 55 (2003) 53–58. disruption in HER2-positive human breast cancer cells, J. Nutr. Biochem. 27
[20] K.M. Rashid, H. Ahsan, S. Siddiqui, W.A. Siddiqui, Tocotrienols have a (2016) 266–277.
nephroprotective action against lipid-induced chronic renal dysfunction in [47] I.G. Algayadh, V. Dronamraju, P.W. Sylvester, Role of Rac1/WAVE2 signaling
rats, Ren. Fail. 37 (2015) 136–143. in mediating the inhibitory effects of ␥-tocotrienol on mammary cancer cell
[21] E.N. Georgousopoulou, D.B. Panagiotakos, D.D. Mellor, N. Naumovski, migration and invasion, Biol. Pharm. Bull. 39 (2016) 1974–1982.
Tocotrienols: health and ageing: a systematic review, Maturitas 95 (2017) [48] F. Khallouki, P. de Medina, S. Caze-Subra, K. Bystricky, P. Balaguer, M. Poirot,
55–60. S. Silvente-Poirot, Molecular and biochemical analysis of the estrogenic and
[22] L. De Silva, L.H. Chuah, P. Meganathan, J.Y. Fu, Tocotrienol and cancer proliferative properties of vitamin E compounds, Front. Oncol. 5 (2016) 287.
metastasis, Biofactors 42 (2016) 149–162. [49] C. Wang, H. Ju, C. Shen, Z. Tong, miR-429 mediates ␦-tocotrienol-induced
[23] S. Wada, Chemoprevention of tocotrienols: the mechanism of apoptosis in triple-negative breast cancer cells by targeting XIAP, Int. J. Clin.
antiproliferative effects, Forum Nutr. 61 (2009) 204–216. Exp. Med. 8 (2015) 15648–15656.
[24] B. Catalgol, S. Batirel, N.K. Ozer, Cellular protection and therapeutic potential [50] W. Gu, I. Prasadam, M. Yu, F. Zhang, P. Ling, Y. Xiao, C. Yu, Gamma
of tocotrienols, Curr. Pharm. Des. 17 (2011) 2215–2220. tocotrienol targets tyrosine phosphatase SHP2 in mammospheres resulting
[25] N. Steuber, K. Vo, R. Wadhwa, J. Birch, P. Iacoban, P. Chavez, T.A. Elbayoumi, in cell death through RAS/ERK pathway, BMC Cancer 15 (2015) 609.
Tocotrienol nanoemulsion platform of curcumin elicit elevated apoptosis [51] P. Parajuli, R.V. Tiwari, P.W. Sylvester, Anti-proliferative effects of
and augmentation of anticancer efficacy against breast and ovarian ␥-tocotrienol are associated with suppression of c-Myc expression in
carcinomas, Int. J. Mol. Sci. 17 (2016) 1792. mammary tumour cells, Cell Prolif. 48 (2015) 421–435.
[26] A. Mahipal, J. Klapman, S. Vignesh, C.S. Yang, A. Neuger, D.T. Chen, M.P. [52] P. Parajuli, R.V. Tiwari, P.W. Sylvester, Anticancer effects of ␥-tocotrienol are
Malafa, Pharmacokinetics and safety of vitamin E ␦-tocotrienol after single associated with a suppression in aerobic glycolysis, Biol. Pharm. Bull. 38
and multiple doses in healthy subjects with measurement of vitamin E (2015) 1352–1360.
metabolites, Cancer Chemother. Pharmacol. 78 (2016) 157–165. [53] R.V. Tiwari, P. Parajuli, P.W. Sylvester, ␥-Tocotrienol-induced autophagy in
[27] G.M. Springett, K. Husain, A. Neuger, B. Centeno, D.T. Chen, T.Z. Hutchinson, malignant mammary cancer cells, Exp. Biol. Med. (Maywood) 239 (2014)
R.M. Lush, S. Sebti, M.P. Malafa, A Phase I Safety, Pharmacokinetic, and 33–44.
270 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

[54] S. Ananthula, P. Parajuli, F.A. Behery, A.Y. Alayoubi, K.A. El Sayed, S. Nazzal, [77] S.J. Shah, P.W. Sylvester, Tocotrienol-induced cytotoxicity is unrelated to
P.W. Sylvester, Oxazine derivatives of ␥- and ␦-tocotrienol display enhanced mitochondrial stress apoptotic signaling in neoplastic mammary epithelial
anticancer activity in vivo, Anticancer Res. 34 (2014) 2715–2726. cells, Biochem. Cell Biol. 83 (2005) 86–95.
[55] R. Loganathan, K.R. Selvaduray, K. Nesaretnam, A.K. Radhakrishnan, [78] S.J. Shah, P.W. Sylvester, Gamma-tocotrienol inhibits neoplastic mammary
Tocotrienols promote apoptosis in human breast cancer cells by inducing epithelial cell proliferation by decreasing Akt and nuclear factor kappaB
poly(ADP-ribose) polymerase cleavage and inhibiting nuclear factor activity, Exp. Biol. Med. (Maywood) 230 (2005) 235–241.
kappa-B activity, Cell Prolif. 46 (2013) 203–213. [79] K. Nesaretnam, R. Ambra, K.R. Selvaduray, A. Radhakrishnan, R. Canali, F.
[56] E. Pierpaoli, V. Viola, A. Barucca, F. Orlando, F. Galli, M. Provinciali, Effect of Virgili, Tocotrienol-rich fraction from palm oil and gene expression in
annatto-tocotrienols supplementation on the development of mammary human breast cancer cells, Ann. N. Y. Acad. Sci. 1031 (2004) 143–157.
tumors in HER-2/neu transgenic mice, Carcinogenesis 34 (6) (2013) [80] K. Nesaretnam, R. Ambra, K.R. Selvaduray, A. Radhakrishnan, K. Reimann, G.
1352–1360. Abdul Razak, F. Virgili, Tocotrienol-rich fraction from palm oil affects gene
[57] P.W. Sylvester, N.M. Ayoub, Tocotrienols target PI3K/Akt signaling in expression in tumors resulting from MCF-7 cell inoculation in athymic mice,
anti-breast cancer therapy, Anticancer Agents Med. Chem. 13 (2013) Lipids 39 (2004) 459–467.
1039–1047. [81] K. Takahashi, G. Loo, Disruption of mitochondria during tocotrienol-induced
[58] V. Viola, S. Ciffolilli, S. Legnaioli, M. Piroddi, M. Betti, F. Mazzini, E. Pierpaoli, apoptosis in MDA-MB-231 human breast cancer cells, Biochem. Pharmacol.
M. Provinciali, F. Galli, Mitochondrial-dependent anticancer activity of 67 (2004) 315–324.
␦-tocotrienol and its synthetic derivatives in HER-2/neu overexpressing [82] S. Shah, P.W. Sylvester, Tocotrienol-induced caspase-8 activation is
breast adenocarcinoma cells, Biofactors 39 (2013) 485–493. unrelated to death receptor apoptotic signaling in neoplastic mammary
[59] D. Patacsil, A.T. Tran, Y.S. Cho, S. Suy, F. Saenz, I. Malyukova, H. Ressom, S.P. epithelial cells, Exp. Biol. Med. (Maywood) 229 (2004) 745–755.
Collins, R. Clarke, D. Kumar, Gamma-tocotrienol induced apoptosis is [83] M.C. Shun, W. Yu, A. Gapor, R. Parsons, J. Atkinson, B.G. Sanders, K. Kline,
associated with unfolded protein response in human breast cancer cells, J. Pro-apoptotic mechanisms of action of a novel vitamin E analog (alpha-TEA)
Nutr. Biochem. 23 (2012) 93–100. and a naturally occurring form of vitamin E (delta-tocotrienol) in
[60] A. Gopalan, W. Yu, Q. Jiang, Y. Jang, B.G. Sanders, K. Kline, Involvement of de MDA-MB-435 human breast cancer cells, Nutr. Cancer 48 (2004) 95–105.
novo ceramide synthesis in gamma-tocopherol and [84] J. Iqbal, M. Minhajuddin, Z.H. Beg, Suppression of 7,12
gamma-tocotrienol-induced apoptosis in human breast cancer cells, Mol. dimethylbenz[alpha]anthracene-induced carcinogenesis and
Nutr. Food Res. 56 (2012) 1803–1811. hypercholesterolaemia in rats by tocotrienol-rich fraction isolated from rice
[61] N.M. Ayoub, S.V. Bachawal, P.W. Sylvester, ␥-Tocotrienol inhibits bran oil, Eur. J. Cancer Prev. 12 (2003) 447–453.
HGF-dependent mitogenesis and Met activation in highly malignant [85] W. Yu, M. Simmons-Menchaca, A. Gapor, B.G. Sanders, K. Kline, Induction of
mammary tumour cells, Cell Prolif. 44 (2011) 516–526. apoptosis in human breast cancer cells by tocopherols and tocotrienols,
[62] P. Ramdas, M. Rajihuzzaman, S.D. Veerasenan, K.R. Selvaduray, K. Nutr. Cancer 33 (1999) 26–32.
Nesaretnam, A.K. Radhakrishnan, Tocotrienol-treated MCF-7 human breast [86] W. Xu, Y. Mi, P. He, S. He, L. Niu, ␥-Tocotrienol inhibits proliferation and
cancer cells show down-regulation of API5 and up-regulation of MIG6 induces apoptosis via the mitochondrial pathway in human cervical cancer
genes, Cancer Genomics Proteomics 8 (2011) 19–31. HeLa cells, Molecules 22 (2017) 1299.
[63] E. Pierpaoli, V. Viola, F. Pilolli, M. Piroddi, F. Galli, M. Provinciali, Gamma- [87] R. Comitato, B. Guantario, G. Leoni, K. Nesaretnam, M.B. Ronci, R. Canali, F.
and delta-tocotrienols exert a more potent anticancer effect than Virgili, Tocotrienols induce endoplasmic reticulum stress and apoptosis in
alpha-tocopheryl succinate on breast cancer cell lines irrespective of cervical cancer cells, Genes Nutr. 11 (2016) 32.
HER-2/neu expression, Life Sci. 86 (2010) 668–675. [88] S. Prasad, S.C. Gupta, A.K. Tyagi, B.B. Aggarwal, ␥-Tocotrienol suppresses
[64] T.C. Hsieh, S. Elangovan, J.M. Wu, Gamma-tocotrienol controls proliferation, growth and sensitises human colorectal tumours to capecitabine in a nude
modulates expression of cell cycle regulatory proteins and up-regulates mouse xenograft model by down-regulating multiple molecules, Br. J.
quinone reductase NQO2 in MCF-7 breast cancer cells, Anticancer Res. 30 Cancer 115 (2016) 814–824.
(2010) 2869–2874. [89] A. Shibata, K. Nakagawa, T. Tsuduki, T. Miyazawa, ␦-Tocotrienol treatment is
[65] R. Comitato, G. Leoni, R. Canali, R. Ambra, K. Nesaretnam, F. Virgili, more effective against hypoxic tumor cells than normoxic cells: potential
Tocotrienols activity in MCF-7 breast cancer cells: involvement of ERbeta implications for cancer therapy, J. Nutr. Biochem. 26 (2015) 832–840.
signal transduction, Mol. Nutr. Food Res. 54 (2010) 669–678. [90] W. Xu, M. Du, Y. Zhao, Q. Wang, W. Sun, B. Chen, ␥-Tocotrienol inhibits cell
[66] W.N. Yap, N. Zaiden, Y.L. Tan, C.P. Ngoh, X.W. Zhang, Y.C. Wong, M.T. Ling, viability through suppression of ␤-catenin/Tcf signaling in human colon
Y.L. Yap, Id1 inhibitor of differentiation, is a key protein mediating carcinoma HT-29 cells, J. Nutr. Biochem. 23 (2012) 800–807.
anti-tumor responses of gamma-tocotrienol in breast cancer cells, Cancer [91] J.S. Zhang, D.M. Li, N. He, Y.H. Liu, C.H. Wang, S.Q. Jiang, B.Q. Chen, J.R. Liu, A
Lett. 291 (2010) 187–199. paraptosis-like cell death induced by ␦-tocotrienol in human colon
[67] T.C. Hsieh, S. Elangovan, J.M. Wu, Differential suppression of proliferation in carcinoma SW620 cells is associated with the suppression of the Wnt
MCF-7 and MDA-MB-231 breast cancer cells exposed to alpha-, gamma- and signaling pathway, Toxicology 11 (1–2) (2011) 8–17.
delta-tocotrienols is accompanied by altered expression of oxidative stress [92] W.L. Xu, J.R. Liu, H.K. Liu, G.Y. Qi, X.R. Sun, W.G. Sun, B.Q. Chen, Inhibition of
modulatory enzymes, Anticancer Res. 30 (2010) 4169–4176. proliferation and induction of apoptosis by gamma-tocotrienol in human
[68] S.K. Park, B.G. Sanders, K. Kline, Tocotrienols induce apoptosis in breast colon carcinoma HT-29 cells, Nutrition 25 (2009) 555–566.
cancer cell lines via an endoplasmic reticulum stress-dependent increase in [93] A. Shibata, K. Nakagawa, P. Sookwong, T. Tsuduki, S. Tomita, H. Shirakawa,
extrinsic death receptor signalling, Breast Cancer Res. Treat. 124 (2010) M. Komai, T. Miyazawa, Tocotrienol inhibits secretion of angiogenic factors
361–375. from human colorectal adenocarcinoma cells by suppressing
[69] K.R. Selvaduray, A.K. Radhakrishnan, M.K. Kutty, K. Nesaretnam, Palm hypoxia-inducible factor-1alpha, J. Nutr. 138 (2008) 2136–2142.
tocotrienols inhibit proliferation of murine mammary cancer cells and [94] A. Shibata, K. Nakagawa, P. Sookwong, T. Tsuduki, S. Oikawa, T. Miyazawa,
induce expression of interleukin-24 mRNA, J. Interferon Cytokine Res. 30 Tumor anti-angiogenic effect and mechanism of action of delta-tocotrienol,
(2010) 909–916. Biochem. Pharmacol. 76 (2008) 330–339.
[70] R. Comitato, K. Nesaretnam, G. Leoni, R. Ambra, R. Canali, A. Bolli, M. Marino, [95] T. Eitsuka, K. Nakagawa, T. Miyazawa, Down-regulation of telomerase
F. Virgili, A novel mechanism of natural vitamin E tocotrienol activity: activity in DLD-1 human colorectal adenocarcinoma cells by tocotrienol,
involvement of ERbeta signal transduction, Am. J. Physiol. Endocrinol. Biochem. Biophys. Res. Commun. 348 (2006) 170–175.
Metab. 297 (2009) E427–437. [96] M.K. Agarwal, M.L. Agarwal, M. Athar, S. Gupta, Tocotrienol-rich fraction of
[71] V.B. Wali, S.V. Bachawal, P.W. Sylvester, Endoplasmic reticulum stress palm oil activates p53, modulates Bax/Bcl2 ratio and induces apoptosis
mediates amma-tocotrienol-induced apoptosis in mammary tumor cells, independent of cell cycle association, Cell Cycle 2 (2004) 205–211.
Apoptosis 14 (2009) 1366–1377. [97] S. Bi, J.R. Liu, Y. Li, Q. Wang, H.K. Liu, Y.G. Yan, B.Q. Chen, W.G. Sun,
[72] W. Weng-Yew, K.R. Selvaduray, C.H. Ming, K. Nesaretnam, Suppression of gamma-Tocotrienol modulates the paracrine secretion of VEGF induced by
tumor growth by palm tocotrienols via the attenuation of angiogenesis, cobalt(II) chloride via ERK signaling pathway in gastric adenocarcinoma
Nutr. Cancer 61 (2009) 367–373. SGC-7901 cell line, Toxicology 274 (2010) 27–33.
[73] S. Elangovan, T.C. Hsieh, J.M. Wu, Growth inhibition of human MDA-mB-231 [98] H.K. Liu, Q. Wang, Y. Li, W.G. Sun, J.R. Liu, Y.M. Yang, W.L. Xu, X.R. Sun, B.Q.
breast cancer cells by delta-tocotrienol is associated with loss of cyclin Chen, Inhibitory effects of gamma-tocotrienol on invasion and metastasis of
D1/CDK4 expression and accompanying changes in the state of human gastric adenocarcinoma SGC-7901 cells, J. Nutr. Biochem. 21 (2010)
phosphorylation of the retinoblastoma tumor suppressor gene product, 206–213.
Anticancer Res. 28 (2008) 2641–2647. [99] Y. Li, W.G. Sun, H.K. Liu, G.Y. Qi, Q. Wang, X.R. Sun, B.Q. Chen, J.R. Liu,
[74] G.V. Samant, P.W. Sylvester, gamma-Tocotrienol inhibits ErbB3-dependent ␥-Tocotrienol inhibits angiogenesis of human umbilical vein endothelial cell
PI3K/Akt mitogenic signalling in neoplastic mammary epithelial cells, Cell induced by cancer cell, J. Nutr. Biochem. 22 (2011) 1127–1136.
Prolif. 39 (2006) 563–574. [100] W. Sun, W. Xu, H. Liu, J. Liu, Q. Wang, J. Zhou, F. Dong, B. Chen,
[75] P.W. Sylvester, S. Shah, Intracellular mechanisms mediating gamma-Tocotrienol induces mitochondria-mediated apoptosis in human
tocotrienol-induced apoptosis in neoplastic mammary epithelial cells, Asia gastric adenocarcinoma SGC-7901 cells, J. Nutr. Biochem. 20 (2009)
Pac. J. Clin. Nutr. 14 (2005) 366–373. 276–284.
[76] P.W. Sylvester, S.J. Shah, G.V. Samant, Intracellular signaling mechanisms [101] W. Sun, Q. Wang, B. Chen, J. Liu, H. Liu, W. Xu, Gamma-tocotrienol-induced
mediating the antiproliferative and apoptotic effects of gamma-tocotrienol apoptosis in human gastric cancer SGC-7901 cells is associated with a
in neoplastic mammary epithelial cells, J. Plant Physiol. 162 (2005) 803–810. suppression in mitogen-activated protein kinase signalling, Br. J. Nutr. 99
(2008) 1247–1254.
B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272 271

[102] S.W. Lim, H.S. Loh, K.N. Ting, T.D. Bradshaw, N.A. Zeenathul, [126] K. Husain, B.A. Centeno, D.T. Chen, W.J. Fulp, M. Perez, G. Zhang Lee, N.
Antiproliferation and induction of caspase-8-dependent Luetteke, S.R. Hingorani, S.M. Sebti, M.P. Malafa, Prolonged survival and
mitochondria-mediated apoptosis by ␤ tocotrienol in human lung and brain delayed progression of pancreatic intraepithelial neoplasia in LSL
cancer cell lines, Biomed. Pharmacother. 68 (2014) 1105–1115. KrasG12D/+;Pdx-1-Cre mice by vitamin E ␦-tocotrienol, Carcinogenesis 34
[103] S.W. Lim, H.S. Loh, K.N. Ting, T.D. Bradshaw, N.A. Zeenathul, Cytotoxicity and (2013) 858–863.
apoptotic activities of alpha-, gamma- and delta-tocotrienol isomers on [127] A.B. Kunnumakkara, B. Sung, J. Ravindran, P. Diagaradjane, A. Deorukhkar, S.
human cancer cells, BMC Complement. Altern. Med. 14 (2014) 469. Dey, C. Koca, V.R. Yadav, Z. Tong, J.G. Gelovani, S. Guha, S. Krishnan, B.B.
[104] A. Inoue, K. Takitani, M. Koh, C. Kawakami, T. Kuno, H. Tamai, Induction of Aggarwal, {Gamma}-tocotrienol inhibits pancreatic tumors and sensitizes
apoptosis by ␥-tocotrienol in human cancer cell lines and leukemic blasts them to gemcitabine treatment by modulating the inflammatory
from patients: dependency on Bid, cytochrome c, and caspase pathway, microenvironment, Cancer Res. 70 (2010) 8695–8705.
Nutr. Cancer 63 (2011) 763–770. [128] D. Hussein, H. Mo, d-␦-Tocotrienol-mediated suppression of the
[105] F. Abdul Rahman Sazli, Z. Jubri, M. Abdul Rahman, S.A. Karsani, A.G. Md Top, proliferation of human PANC-1, MIA PaCa-2, and BxPC-3 pancreatic
W.Z. Wan Ngah, Gamma-tocotrienol treatment increased peroxiredoxin-4 carcinoma cells, Pancreas 38 (2009) e124–e136.
expression in HepG2 liver cancer cell line, BMC Complement. Altern. Med. [129] N. Shiozawa, R. Sugahara, K. Namiki, C. Sato, A. Ando, A. Sato, N. Virgona, T.
(2015) 15–64. Yano, Inhibitory effect of a redox-silent analogue of tocotrienol on hypoxia
[106] K.S. Siveen, K.S. Ahn, T.H. Ong, M.K. Shanmugam, F. Li, W.N. Yap, A.P. Kumar, adaptation in prostate cancer cells, Anticancer Drugs 28 (2017) 289–297.
C.W. Fong, V. Tergaonkar, K.M. Hui, G. Sethi, Y-tocotrienol inhibits [130] Y. Huang, R. Wu, Z.Y. Su, Y. Guo, X. Zheng, C.S. Yang, A.N. Kong, A naturally
angiogenesis-dependent growth of human hepatocellular carcinoma occurring mixture of tocotrienols inhibits the growth of human prostate
through abrogation of AKT/mTOR pathway in an orthotopic mouse model, tumor, associated with epigenetic modifications of cyclin-dependent kinase
Oncotarget 5 (2014) 1897–1911. inhibitors p21 and p27, J. Nutr. Biochem. 40 (2017) 155–163.
[107] P. Rajendran, F. Li, K.A. Manu, M.K. Shanmugam, S.Y. Loo, A.P. Kumar, G. [131] R. Sugahara, A. Sato, A. Uchida, S. Shiozawa, C. Sato, N. Virgona, T. Yano,
Sethi, ␥-Tocotrienol is a novel inhibitor of constitutive and inducible STAT3 Annatto tocotrienol induces a cytotoxic effect on human prostate cancer
signalling pathway in human hepatocellular carcinoma: potential role as an PC3Cells via the simultaneous inhibition of src and stat3, J. Nutr. Sci.
antiproliferative, pro-apoptotic and chemosensitizing agent, Br. J. Vitaminol. (Tokyo) 61 (2015) 497–501.
Pharmacol. 163 (2011) 283–298. [132] Q. Jiang, X. Rao, C.Y. Kim, H. Freiser, Q. Zhang, Z. Jiang, G. Li, Gamma
[108] N. Zaiden, W.N. Yap, S. Ong, C.H. Xu, V.H. Teo, C.P. Chang, X.W. Zhang, K. tocotrienol induces apoptosis and autophagy in prostate cancer cells by
Nesaretnam, S. Shiba, Y.L. Yap, Gamma delta tocotrienols reduce hepatic increasing intracellular dihydrosphingosine and dihydroceramide, Int. J.
triglyceride synthesis and VLDL secretion, J. Atheroscler. Thromb. 17 (2010) Cancer 130 (2012) 685–693.
1019–1032. [133] J.R. Krycer, L. Phan, A.J. Brown, A key regulator of cholesterol homoeostasis
[109] Y. Hiura, H. Tachibana, R. Arakawa, N. Aoyama, M. Okabe, M. Sakai, K. SREBP-2, can be targeted in prostate cancer cells with natural products,
Yamada, Specific accumulation of gamma- and delta-tocotrienols in tumor Biochem. J. 446 (2012) 191–201.
and their antitumor effect in vivo, J. Nutr. Biochem. 20 (2009) 607–613. [134] S.E. Campbell, B. Rudder, R.B. Phillips, S.G. Whaley, J.B. Stimmel, L.M.
[110] M. Sakai, M. Okabe, H. Tachibana, K. Yamada, Apoptosis induction by Leesnitzer, J. Lightner, S. Dessus-Babus, M. Duffourc, W.L. Stone, D.G.
gamma-tocotrienol in human hepatoma Hep3B cells, J. Nutr. Biochem. 17 Menter, R.A. Newman, P. Yang, B.B. Aggarwal, K. Krishnan, ␥-Tocotrienol
(2006) 672–676. induces growth arrest through a novel pathway with TGF␤2 in prostate
[111] S. Wada, Y. Satomi, M. Murakoshi, N. Noguchi, T. Yoshikawa, H. Nishino, cancer, Free Radic. Biol. Med. 50 (2011) 1344–1354.
Tumor suppressive effects of tocotrienol in vivo and in vitro, Cancer Lett. [135] S.U. Luk, W.N. Yap, Y.T. Chiu, D.T. Lee, S. Ma, T.K. Lee, R.S. Vasireddy, Y.C.
229 (2) (2005) 181–191. Wong, Y.P. Ching, C. Nelson, Y.L. Yap, M.T. Ling, Gamma-tocotrienol as an
[112] C.H. Har, C.K. Keong, Effects of tocotrienols on cell viability and apoptosis in effective agent in targeting prostate cancer stem cell-like population, Int. J.
normal murine liver cells (BNL CL.2) and liver cancer cells (BNL 1ME A.7R.1), Cancer 128 (2011) 2182–2191.
in vitro, Asia Pac. J. Clin. Nutr. 14 (2005) 374–380. [136] W.N. Yap, P.N. Chang, H.Y. Han, D.T. Lee, M.T. Ling, Y.C. Wong, Y.L. Yap,
[113] M. Sakai, M. Okabe, M. Yamasaki, H. Tachibana, K. Yamada, Induction of Gamma-tocotrienol suppresses prostate cancer cell proliferation and
apoptosis by tocotrienol in rat hepatoma dRLh-84 cells, Anticancer Res. 24 invasion through multiple-signalling pathways, Br. J. Cancer 99 (2008)
(2004) 1683–1688. 1832–1841.
[114] A. Rahmat, W.Z. Ngah, N.A. Shamaan, A. Gapor, K. Abdul Kadir, Long-term [137] J.K. Srivastava, S. Gupta, Tocotrienol-rich fraction of palm oil induces cell
administration of tocotrienols and tumor-marker enzyme activities during cycle arrest and apoptosis selectively in human prostate cancer cells,
hepatocarcinogenesis in rats, Nutrition 9 (1993) 229–232. Biochem. Biophys. Res. Commun. 346 (2006) 447–453.
[115] W.Z. Ngah, Z. Jarien, M.M. San, A. Marzuki, G.M. Top, N.A. Shamaan, K.A. [138] M. Montagnani Marelli, M. Marzagalli, R.M. Moretti, G. Beretta, L. Casati, R.
Kadir, Effect of tocotrienols on hepatocarcinogenesis induced by Comitato, G.L. Gravina, C. Festuccia, P. Limonta, E. Vitamin, Vitamin E
2-acetylaminofluorene in rats, Am. J. Clin. Nutr. 53 (1991) 1076S–1081S. ␦-tocotrienol triggers endoplasmic reticulum stress-mediated apoptosis in
[116] X. Ji, Z. Wang, A. Geamanu, A. Goja, F.H. Sarkar, S.V. Gupta, Delta-tocotrienol human melanoma cells, Sci. Rep. 6 (2016) 30502.
suppresses Notch-1 pathway by upregulating miR-34a in non-small cell [139] L.T. Ng, L.T. Lin, C.L. Chen, H.W. Chen, S.J. Wu, C.C. Lin, Anti-melanogenic
lung cancer cells, Int. J. Cancer 131 (2012) 2668–2677. effects of ␦-tocotrienol are associated with tyrosinase-related proteins and
[117] K. Kashiwagi, K. Harada, Y. Yano, I. Kumadaki, K. Hagiwara, J. Takebayashi, MAPK signaling pathway in B16 melanoma cells, Phytomedicine 21 (2014)
W. Kido, N. Virgona, T. Yano, A redox-silent analogue of tocotrienol inhibits 978–983.
hypoxic adaptation of lung cancer cells, Biochem. Biophys. Res. Commun. [140] B. Choi, J.H. Heo, H.J. Kwon, E.S. Lee, S. Sohn, Tocotrienols enhance
365 (2008) 875–881. melanosome degradation through endosome docking/fusion proteins in
[118] Y. Yano, H. Satoh, K. Fukumoto, I. Kumadaki, T. Ichikawa, K. Yamada, K. B16F10 melanoma cells, Food Funct. 4 (2013) 1481–1488.
Hagiwara, T. Yano, Induction of cytotoxicity in human lung adenocarcinoma [141] N.V. Fernandes, P.K. Guntipalli, H. Mo, d-␦-Tocotrienol-mediated cell cycle
cells by 6-O-carboxypropyl-alpha-tocotrienol, a redox-silent derivative of arrest and apoptosis in human melanoma cells, Anticancer Res. 30 (2010)
alpha-tocotrienol, Int. J. Cancer 115 (2005) 839–846. 4937–4944.
[119] A. Sato, H. Ueno, A. Takase, A. Ando, Y. Sekine, T. Yano, Cytotoxicity induced [142] P.N. Chang, W.N. Yap, D.T. Lee, M.T. Ling, Y.C. Wong, Y.L. Yap, Evidence of
by a redox-silent analog of tocotrienol in human mesothelioma H2452 cell gamma-tocotrienol as an apoptosis-inducing, invasion-suppressing, and
line via suppression of cap-dependent protein translation, Anticancer Res. chemotherapy drug-sensitizing agent in human melanoma cells, Nutr.
36 (2016) 1527–1533. Cancer 61 (2009) 357–366.
[120] A. Sato, N. Virgona, A. Ando, M. Ota, T. Yano, A redox-silent analogue of [143] A. Xiong, W. Yu, Y. Liu, B.G. Sanders, K. Kline, Elimination of ALDH+ breast
tocotrienol inhibits cobalt (II) chloride-induced VEGF expression via Yes tumor initiating cells by docosahexanoic acid and/or gamma tocotrienol
signaling in mesothelioma cells, Biol. Pharm. Bull. 37 (2014) 865–870. through SHP-1 inhibition of Stat3 signaling, Mol. Carcinog. 55 (2016)
[121] K. Kashiwagi, N. Virgona, K. Harada, W. Kido, Y. Yano, A. Ando, K. Hagiwara, 420–430.
T. Yano, A redox-silent analogue of tocotrienol acts as a potential cytotoxic [144] A.T. Tran, M. Ramalinga, H. Kedir, R. Clarke, D. Kumar, Autophagy inhibitor
agent against human mesothelioma cells, Life Sci. 84 (2009) 650–656. 3-methyladenine potentiates apoptosis induced by dietary tocotrienols in
[122] J.K. Tan, S.M. Then, M. Mazlan, R.N. Raja Abdul Rahman, R. Jamal, W.Z. Wan breast cancer cells, Eur. J. Nutr. 54 (2015) 265–272.
Ngah, Gamma-tocotrienol acts as a BH3 mimetic to induce apoptosis in [145] T. Eitsuka, N. Tatewaki, H. Nishida, T. Kurata, K. Nakagawa, T. Miyazawa,
neuroblastoma SH-SY5Y cells, J. Nutr. Biochem. 31 (2016) 28–37. Synergistic inhibition of cancer cell proliferation with a combination of
[123] K. Husain, B.A. Centeno, D. Coppola, J. Trevino, S.M. Sebti, M.P. Malafa, (-tocotrienol and ferulic acid, Biochem. Biophys. Res. Commun. 453 (2014)
␦-Tocotrienol, a natural form of vitamin E, inhibits pancreatic cancer 606–611.
stem-like cells and prevents pancreatic cancer metastasis, Oncotarget 8 [146] A. Malaviya, P.W. Sylvester, Synergistic antiproliferative effects of combined
(2017) 31554–31567. ␥-tocotrienol and PPAR ␥ antagonist treatment are mediated through PPAR
[124] C. Wang, K. Husain, A. Zhang, B.A. Centeno, D.T. Chen, Z. Tong, S.M. Sebti, M.P. ␥-Independent mechanisms in breast cancer cells, PPAR Res. 2014 (2014)
Malafa, EGR-1/Bax pathway plays a role in vitamin E ␦-tocotrienol-induced 439146.
apoptosis in pancreatic cancer cells, J. Nutr. Biochem. 26 (2015) 797–807. [147] N.M. Ayoub, M.R. Akl, P.W. Sylvester, Combined ␥-tocotrienol and Met
[125] P.J. Hodul, Y. Dong, K. Husain, J.M. Pimiento, J. Chen, A. Zhang, R. Francois, inhibitor treatment suppresses mammary cancer cell proliferation,
W.J. Pledger, D. Coppola, S.M. Sebti, D.T. Chen, M.P. Malafa, Vitamin E. epithelial-to-mesenchymal transition and migration, Cell Prolif. 46 (2013)
␦-tocotrienol induces p27 (Kip1)-dependent cell-cycle arrest in pancreatic 538–553.
cancer cells via an E2F-1-dependent mechanism, PLoS One 8 (2013) e52526.
272 B.L. Sailo et al. / Pharmacological Research 130 (2018) 259–272

[148] S.R. Abdul Hafid, S. Chakravarthi, K. Nesaretnam, A.K. Radhakrishnan, [168] M. Nasr, N. Nafee, H. Saad, A. Kazem, Improved antitumor activity and
Tocotrienol-adjuvanted dendritic cells inhibit tumor growth and reduced cardiotoxicity of epirubicin using hepatocyte-targeted
metastasis: a murine model of breast cancer, PLoS One 8 (2013) e74753. nanoparticles combined with tocotrienols against hepatocellular carcinoma
[149] M.R. Akl, N.M. Ayoub, B.S. Abuasal, A. Kaddoumi, P.W. Sylvester, Sesamin in mice, Eur. J. Pharm. Biopharm. 88 (2014) 216–225.
synergistically potentiates the anticancer effects of ␥-tocotrienol in [169] J.A. McAnally, J. Gupta, S. Sodhani, L. Bravo, H. Mo, Tocotrienols potentiate
mammary cancer cell lines, Fitoterapia 84 (2013) 347–359. lovastatin-mediated growth suppression in vitro and in vivo, Exp. Biol. Med.
[150] A. Malaviya, P.W. Sylvester, Mechanisms mediating the effects of (Maywood) 232 (2007) 523–531.
␥-tocotrienol when used in combination with PPAR␥ agonists or [170] K. Nakashima, N. Virgona, M. Miyazawa, T. Watanabe, T. Yano, The
antagonists on MCF-7 and MDA-MB-231 Breast cancer cells, Int. J. Breast tocotrienol-rich fraction from rice bran enhances cisplatin-induced
Cancer 2013 (2013) 101705. cytotoxicity in human mesothelioma H28 cells, Phytother. Res. 24 (2010)
[151] P.W. Sylvester, Synergistic anticancer effects of combined ␥-tocotrienol 1317–1321.
with statin or receptor tyrosine kinase inhibitor treatment, Genes Nutr. 7 [171] G. Tuerdi, S. Ichinomiya, H. Sato, S. Siddig, E. Suwa, H. Iwata, T. Yano, K. Ueno,
(2012) 63–74. Synergistic effect of combined treatment with gamma-tocotrienol and statin
[152] A. Gopalan, W. Yu, B.G. Sanders, K. Kline, Eliminating drug resistant breast on human malignant mesothelioma cells, Cancer Lett. 339 (2013) 116–127.
cancer stem-like cells with combination of simvastatin and gamma [172] K. Kani, Y. Momota, M. Harada, Y. Yamamura, K. Aota, T. Yamanoi, H. Takano,
tocotrienol, Cancer Lett. 328 (2013) 285–296. K. Motegi, M. Azuma, ␥-tocotrienol enhances the chemosensitivity of
[153] M.R. Akl, N.M. Ayoub, P.W. Sylvester, Mechanisms mediating the synergistic human oral cancer cells to docetaxel through the downregulation of the
anticancer effects of combined ␥-tocotrienol and sesamin treatment, Planta expression of NF-␬B-regulated anti-apoptotic gene products, Int. J. Oncol. 42
Med. 78 (16) (2012) 1731–1739. (2013) 75–82.
[154] N.M. Ayoub, S.V. Bachawal, P.W. Sylvester, ␥-Tocotrienol inhibits [173] K. Husain, R.A. Francois, T. Yamauchi, M. Perez, S.M. Sebti, M.P. Malafa, E.
HGF-dependent mitogenesis and Met activation in highly malignant Vitamin, ␦-tocotrienol augments the antitumor activity of gemcitabine and
mammary tumour cells, Cell Prolif. 44 (2011) 516–526. suppresses constitutive NF-␬B activation in pancreatic cancer, Mol. Cancer
[155] A. Barve, T.O. Khor, K. Reuhl, B. Reddy, H. Newmark, A.N. Kong, Mixed Ther. 10 (2011) 2363–2372.
tocotrienols inhibit prostate carcinogenesis in TRAMP mice, Nutr. Cancer 62 [174] J. Liu, E.Y. Lau, J. Chen, J. Yong, K.D. Tang, J. Lo, I.O. Ng, T.K. Lee, M.T. Ling,
(2010) 789–794. Polysaccharopeptide enhanced the anti-cancer effect of gamma-tocotrienol
[156] A.B. Shirode, P.W. Sylvester, Synergistic anticancer effects of combined through activation of AMPK, BMC Complement. Altern. Med. 14 (2014) 303.
gamma-tocotrienol and celecoxib treatment are associated with [175] W.N. Yap, N. Zaiden, S.Y. Luk, D.T. Lee, M.T. Ling, Y.C. Wong, Y.L. Yap, In vivo
suppression in Akt and NFkappaB signaling, Biomed. Pharmacother. 64 evidence of gamma-tocotrienol as a chemosensitizer in the treatment of
(2010) 327–332. hormone-refractory prostate cancer, Pharmacology 85 (2010) 248–258.
[157] S.V. Bachawal, V.B. Wali, P.W. Sylvester, Enhanced antiproliferative and [176] K. Nesaretnam, K.R. Selvaduray, G. Abdul Razak, S.D. Veerasenan, P.A.
apoptotic response to combined treatment of gamma-tocotrienol with Gomez, Effectiveness of tocotrienol-rich fraction combined with tamoxifen
erlotinib or gefitinib in mammary tumor cells, BMC Cancer 8 (2010) 10–84. in the management of women with early breast cancer: a pilot clinical trial,
[158] S.V. Bachawal, V.B. Wali, P.W. Sylvester, Combined gamma-tocotrienol and Breast Cancer Res. 12 (2010) R81.
erlotinib/gefitinib treatment suppresses Stat and Akt signaling in murine [177] Clinicaltrials.gov, US National Library of Medicine. https://clinicaltrials.gov/,
mammary tumor cells, Anticancer Res. 30 (2010) 429–437. (Accessed on 9, November 2017).
[159] V.B. Wali, S.V. Bachawal, P.W. Sylvester, Combined treatment of [178] D.C. Schwenke, Does lack of tocopherols and tocotrienols put women at
gamma-tocotrienol with statins induce mammary tumor cell cycle arrest in increased risk of breast cancer, J. Nutr. Biochem. 13 (2002) 2–20.
G1, Exp. Biol. Med. (Maywood) 234 (2009) 639–650. [179] K. Nesaretnam, N. Guthrie, A.F. Chambers, K.K. Carroll, Effect of tocotrienols
[160] T.C. Hsieh, J.M. Wu, Suppression of cell proliferation and gene expression by on the growth of a human breast cancer cell line in culture, Lipids 30 (1995)
combinatorial synergy of EGCG, resveratrol and gamma-tocotrienol in 1139–1143.
estrogen receptor-positive MCF-7 breast cancer cells, Int. J. Oncol. 33 (2008) [180] K. Nesaretnam, R. Stephen, R. Dils, P. Darbre, Tocotrienols inhibit the growth
851–859. of human breast cancer cells irrespective of estrogen receptor status, Lipids
[161] V.B. Wali, P.W. Sylvester, Synergistic antiproliferative effects of 33 (1998) 461–469.
gamma-tocotrienol and statin treatment on mammary tumor cells, Lipids [181] K. Nesaretnam, S. Dorasamy, P.D. Darbre, Tocotrienols inhibit growth of
42 (2007) 1113–1123. ZR-75-1 breast cancer cells, Int. J. Food Sci. Nutr. 51 (2000) 95–103.
[162] I.B. Abubakar, K.H. Lim, T.S. Kam, H.S. Loh, Synergistic cytotoxic effects of [182] P.W. Sylvester, A. Nachnani, S. Shah, K.P. Briski, Role of GTP-binding proteins
combined ␦-tocotrienol and jerantinine B on human brain and colon in reversing the antiproliferative effects of tocotrienols in preneoplastic
cancers, J. Ethnopharmacol. 184 (2016) 107–118. mammary epithelial cells, Asia Pac. J. Clin. Nutr. 11 (2002) S452–S459.
[163] Z. Yang, M.J. Lee, Y. Zhao, C.S. Yang, Metabolism of tocotrienols in animals [183] R.V. Tiwari, P. Parajuli, P.W. Sylvester, ␥-Tocotrienol-induced autophagy in
and synergistic inhibitory actions of tocotrienols with atorvastatin in cancer malignant mammary cancer cells, Exp. Biol. Med. (Maywood) 239 (2014)
cells, Genes Nutr. 7 (2012) 11–18. 33–44.
[164] Z. Yang, H. Xiao, H. Jin, P.T. Koo, D.J. Tsang, C.S. Yang, Synergistic actions of [184] M.L. de Mesquita, R.M. Araújo, D.P. Bezerra, R.B. Filho, J.E. de Paula, E.R.
atorvastatin with gamma-tocotrienol and celecoxib against human colon Silveira, C. Pessoa, M.O. de Moraes, L.V. Costa Lotufo, L.S. Espindola,
cancer HT29 and HCT116 cells, Int. J. Cancer 126 (2010) 852–863. Cytotoxicity of ␦-tocotrienols from Kielmeyera coriacea against cancer cell
[165] K.A. Manu, M.K. Shanmugam, L. Ramachandran, F. Li, C.W. Fong, A.P. Kumar, lines, Bioorg. Med. Chem. 19 (2011) 623–630.
P. Tan, G. Sethi, First evidence that ␥-tocotrienol inhibits the growth of [185] J.Y. Fu, D.R. Blatchford, L. Tetley, C. Dufès, Tumor regression after systemic
human gastric cancer and chemosensitizes it to capecitabine in a xenograft administration of tocotrienol entrapped in tumor-targeted vesicles, J.
mouse model through the modulation of NF-␬B pathway, Clin. Cancer Res. Control. Release 140 (2009) 95–99.
18 (2012) 2220–2229. [186] G.C. Burdeos, K. Nakagawa, A. Watanabe, F. Kimura, T. Miyazawa,
[166] A. Abdul Rahman, A.R. Jamal, R. Harun, N. Mohd Mokhtar, W.Z. Wan Ngah, (-Tocotrienol attenuates triglyceride through effect on lipogenic gene
Gamma-tocotrienol and hydroxy-chavicol synergistically inhibits growth expressions in mouse hepatocellular carcinoma Hepa 1–6, J. Nutr. Sci.
and induces apoptosis of human glioma cells, BMC Complement. Altern. Vitaminol. (Tokyo) 59 (2013) 148–151.
Med. 14 (2014) 213. [187] R. Karim, S. Somani, M. Al Robaian, M. Mullin, R. Amor, G. McConnell, C.
[167] C.C. Chen, T.Y. Liu, S.P. Huang, C.T. Ho, T.C. Huang, Differentiation and Dufès, Tumor regression after intravenous administration of targeted
apoptosis induction by lovastatin and ␥-tocotrienol in HL-60 cells via vesicles entrapping the vitamin E ␣-tocotrienol, J. Control. Release 246
Ras/ERK/NF-␬B and Ras/Akt/NF-␬B signaling dependent down-regulation of (2017) 79–87.
glyoxalase 1 and HMG-CoA reductase, Cell. Signal. 27 (2015) 2182–2190. [188] R. Roskoski Jr., Guidelines for preparing color figures for everyone including
the colorblind, Pharmacol. Res. 119 (2017) 240–241.

Vous aimerez peut-être aussi